handbook of marine natural products volume 426 || enzyme inhibitors from marine invertebrates

86
Enzyme Inhibitors from Marine Invertebrates 23 Yoichi Nakao and Nobuhiro Fusetani Contents 23.1 Introduction ............................................................................. 1146 23.2 Oxidoreductases (EC 1.-.-.-) ........................................................... 1147 23.2.1 Aldose Reductase (EC 1.1.1.21) .............................................. 1147 23.2.2 Enoyl-ACP Reductase (EC 1.3.1.9) .......................................... 1148 23.2.3 Lipoxygenases (EC 1.13.11.12) .............................................. 1149 23.2.4 Indoleamine 2,3-Dioxygenases (EC 1.13.11.42) ............................ 1150 23.2.5 Neuronal Nitric Oxide Synthase (EC 1.14.13.39) ........................... 1151 23.3 Transferases (EC 2.-.-.-) ................................................................ 1152 23.3.1 1a-1,3-Fucosyltransferases (EC 2.4.1.214) ................................. 1152 23.3.2 Adenosine Phosphoribosyltransferase (EC 2.4.2.7) ........................ 1153 23.3.3 Farnesyl Protein Transferase (EC 2.5.1.58) ................................ 1154 23.3.4 Geranylgeranyltransferase Type I (EC 2.5.1.59) ........................... 1156 23.3.5 HIV Reverse Transcriptase (EC 2.7.7.49) .................................. 1157 23.3.6 HIV Integrase (EC 2.7.7.-) .................................................. 1158 23.3.7 Telomerase (EC 2.7.7.49) ................................................... 1160 23.3.8 Epidermal Growth Factor Receptor Kinase (EC 2.7.10.1) ................. 1162 23.3.9 Tyrosine Kinase pp60 V-SRC (EC 2.7.10.2) .................................. 1163 23.3.10 Other Tyrosine Protein Kinase .............................................. 1164 23.3.11 MSK1 (EC 2.7.11.1) and MAPKAPK-2 .................................... 1164 23.3.12 Raf/MEK1/MAPK ........................................................... 1164 23.3.13 Checkpoint Kinases (EC 2.7.11.1) .......................................... 1164 23.3.14 Protein Kinase C (EC 2.7.11.13) ............................................ 1166 23.3.15 Cyclin-Dependent Kinases (EC 2.7.11.22) ................................. 1170 23.3.16 Glycogen Synthase Kinase-3 (2.7.11.17) ................................... 1171 23.4 Hydrolases (EC 3.-.-.-) ................................................................. 1172 23.4.1 Phospholipase A 2 (EC 3.1.1.4) .............................................. 1172 Y. Nakao (*) School of Advanced Sciences and Technologies, Waseda University, 3-4-1, Okubo, Shinjuku-ku, Tokyo, Japan N. Fusetani Fisheries and Oceans Hakodate, Graduate School of Fisheries Sciences, Hokkaido University, 3-1-1, Minato-cho, Hakodate, Japan E. Fattorusso, W. H. Gerwick, O. Taglialatela-Scafati (eds.), Handbook of Marine Natural Products, DOI 10.1007/978-90-481-3834-0_23, # Springer Science+Business Media B.V. 2012 1145

Upload: orazio

Post on 08-Oct-2016

237 views

Category:

Documents


15 download

TRANSCRIPT

Page 1: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

Enzyme Inhibitors from MarineInvertebrates 23Yoichi Nakao and Nobuhiro Fusetani

Contents

23.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1146

23.2 Oxidoreductases (EC 1.-.-.-) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1147

23.2.1 Aldose Reductase (EC 1.1.1.21) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1147

23.2.2 Enoyl-ACP Reductase (EC 1.3.1.9) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1148

23.2.3 Lipoxygenases (EC 1.13.11.12) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1149

23.2.4 Indoleamine 2,3-Dioxygenases (EC 1.13.11.42) . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1150

23.2.5 Neuronal Nitric Oxide Synthase (EC 1.14.13.39) . . . . . . . . . . . . . . . . . . . . . . . . . . . 1151

23.3 Transferases (EC 2.-.-.-) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1152

23.3.1 1a-1,3-Fucosyltransferases (EC 2.4.1.214) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1152

23.3.2 Adenosine Phosphoribosyltransferase (EC 2.4.2.7) . . . . . . . . . . . . . . . . . . . . . . . . 1153

23.3.3 Farnesyl Protein Transferase (EC 2.5.1.58) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1154

23.3.4 Geranylgeranyltransferase Type I (EC 2.5.1.59) . . . . . . . . . . . . . . . . . . . . . . . . . . . 1156

23.3.5 HIV Reverse Transcriptase (EC 2.7.7.49) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1157

23.3.6 HIV Integrase (EC 2.7.7.-) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1158

23.3.7 Telomerase (EC 2.7.7.49) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1160

23.3.8 Epidermal Growth Factor Receptor Kinase (EC 2.7.10.1) . . . . . . . . . . . . . . . . . 1162

23.3.9 Tyrosine Kinase pp60V-SRC (EC 2.7.10.2) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1163

23.3.10 Other Tyrosine Protein Kinase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1164

23.3.11 MSK1 (EC 2.7.11.1) and MAPKAPK-2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1164

23.3.12 Raf/MEK1/MAPK . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1164

23.3.13 Checkpoint Kinases (EC 2.7.11.1) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1164

23.3.14 Protein Kinase C (EC 2.7.11.13) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1166

23.3.15 Cyclin-Dependent Kinases (EC 2.7.11.22) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1170

23.3.16 Glycogen Synthase Kinase-3 (2.7.11.17) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1171

23.4 Hydrolases (EC 3.-.-.-) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1172

23.4.1 Phospholipase A2 (EC 3.1.1.4) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1172

Y. Nakao (*)

School of Advanced Sciences and Technologies, Waseda University, 3-4-1, Okubo, Shinjuku-ku,

Tokyo, Japan

N. Fusetani

Fisheries and Oceans Hakodate, Graduate School of Fisheries Sciences, Hokkaido University,

3-1-1, Minato-cho, Hakodate, Japan

E. Fattorusso, W. H. Gerwick, O. Taglialatela-Scafati (eds.),

Handbook of Marine Natural Products, DOI 10.1007/978-90-481-3834-0_23,# Springer Science+Business Media B.V. 2012

1145

Page 2: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

23.4.2 Acetylcholinesterase (EC 3.1.1.7) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1177

23.4.3 Protein Phosphatases (EC 3.1.3.16) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1178

23.4.4 Protein Tyrosine Phosphatases (EC 3.1.3.48) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1183

23.4.5 Phospholipase C (EC 3.1.4.3) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1184

23.4.6 Phosphodiesterase (EC 3.1.4.-) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1185

23.4.7 Sialidase (EC 3.2.1.18) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1186

23.4.8 Chitinase (EC 3.2.1.14) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1188

23.4.9 a-Glucosidases (EC 3.2.1.20) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1189

23.4.10 S-Adenosylhomocysteine Hydrolase (EC 3.3.1.1) . . . . . . . . . . . . . . . . . . . . . . . . 1190

23.4.11 Serine Proteases (EC 3.4.21.-) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1191

23.4.12 Cysteine Proteases (EC 3.4.22.-) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1196

23.4.13 Aspartic Proteases (EC 3.4.23.-) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1198

23.4.14 Metalloproteases (EC 3.4.24.-) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1198

23.4.15 Histone Deacetylases (EC 3.23.-) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1200

23.4.16 ATPases (EC 3.6.1.3) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1202

23.5 Lyases (EC 4.-.-.-) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1208

23.5.1 ATP Citrate Lyase (EC 4.1.3.8) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1208

23.6 Isomerases (EC 5.-.-.-) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1209

23.6.1 Topoisomerase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1209

23.7 Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1213

23.8 Study Questions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1213

References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1214

Abstract

Marine invertebrates are the rich source of small molecules with unique

chemical skeletons and potent bioactivities; however, the major biological

activities tested for marine natural products have been mainly limited to

the traditional phenotype-oriented bioactivities such as cytotoxicity or anti-

microbial activities. Enzyme inhibition is the target-oriented bioactivity and

small molecule inhibitors are quite useful for controlling biological processes

and understanding complicated biological phenomena. Recently, it has been

recognized that the natural products are superior source of small molecule

probes useful for the chemical biology. Especially, marine-derived enzyme

inhibitors still remain to be fully investigated in spite of their high potentials

as a source of unique compounds. In this chapter, selected enzyme inhibitors

from marine invertebrates are described.

23.1 Introduction

Enzymes are vital to living organisms, maintaining the homeostasis of their lives by

mediating/regulating numerous biochemical events, including metabolism, catab-

olism, cellular signal transduction, cell cycles, and development. Dysfunction,

overexpression, or hyperactivation of the enzymes are often associated with

human diseases as evidenced from the molecular-based analysis of diseases.

Umezawa’s pioneering work on enzyme inhibitors resulted in the discovery of

1146 Y. Nakao and N. Fusetani

Page 3: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

numbers of valuable compounds from terrestrial microorganisms [1]. Thereafter,

search for small molecular enzyme inhibitors has been actively conducted in both

academia and pharmaceutical industries, and a considerable number of enzyme

inhibitors have been developed as drugs, including blockbusters such as statins.

However, only recently, the majority of the marine natural products chemists

started systematic programs for discovering enzyme inhibitors; the first systematic

investigation was done by our group for H,K-ATPase inhibitors [2]. Although

a large proportion of enzyme inhibitors reported from marine organisms were

known compounds found to inhibit enzymes in the reevaluation of activities,

these known compounds with new activities are important for the related research

areas such as chemical biology. It may be predicted that the trends of marine natural

products chemistry will move from finding merely “new structures” to finding “newactivities” of known compounds. Recent reevaluation of natural products as

a superior source of small molecule probes useful for studying biological phenom-

ena is encouraging natural products chemists to investigate compounds with new

activities.

In this chapter, we will describe the structures and activities of selected

enzyme inhibitors isolated from marine invertebrates. Some inhibitors are not

from marine invertebrates, but they are related to structures from microorgan-

isms. Synthetic enzyme inhibitors based on the marine natural products are also

described. Most of them were designed based on the crystal structures of

enzyme–inhibitor complexes and gave a detailed insight into the modes of

action. However, we excluded most of bromotyrosines, polyacetylenes, and

highly sulfated steroids which are often termed “nuisance compounds,” because

they show a variety of biological activities including inhibition of enzymes.

Inhibitors are listed according to the classification of target enzymes

(EC number).

23.2 Oxidoreductases (EC 1.-.-.-)

23.2.1 Aldose Reductase (EC 1.1.1.21)

Aldose reductase catalyzes the reduction of glucose to sorbitol with concomitant

conversion of NADPH to NAPD+. The highly activated enzyme under hypergly-

cemic environment results in unusual accumulation of sorbitol in the eye lens or

peripheral nerves that lead to retinopathy, cataract, neuropathy, nephropathy, or

cardio-vascular diseases. Three polycyclic bis-indoles (1–3) from a marine sponge

Dictyodendrilla sp. strongly inhibited bovine lens aldose reductase with IC50 values

of 49, 125, and 112 nM, respectively. Interestingly, the sulfate group in 1 and 2 didnot influence the activity (4: IC50 102 nM), while the sulfate group in 3 potentiatedthe activity (5: IC50 567 nM) [3].

23 Enzyme Inhibitors from Marine Invertebrates 1147

Page 4: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

N

NH

O

OR

OH

OH

HO

O

HO

HO

O

N

NH

O

OR

OH

OH

HO

MeOOC

HO

1 R = SO3Na2 R = SO3H4 R = H

3 R = SO3H5 R = H

23.2.2 Enoyl-ACP Reductase (EC 1.3.1.9)

In mammals and other higher eukaryotes, all reactions in fatty acid synthesis are

catalyzed by the type I fatty acid synthase (FAS-I), while bacteria, plants, and algae

contain a type II system (FAS-II), in which each reaction is carried out by

a monofunctional enzyme. The type II fatty acid biosynthetic pathway has recently

been discovered in a number of Apicomplexan parasites, including the malaria

parasite, Plasmodium falciparum. Mycobacterium tuberculosis employs both

FAS-I and FAS-II systems. FAS-I is responsible for the synthesis of short-chain

FAs, whereas the FAS-II system extends these FAs to very long-chain

mycolic acids, important components of the mycobacterial cell wall. The central

role of FA biosynthesis and the structural differences between the human and

microbial FAS systems render FAS-II as an attractive target for antimicrobial

drug discovery.

The FabI (enoyl-ACP reductase) is a crucial enzyme of all FAS-II systems, since it

catalyzes the last NADH-dependent reduction step in each elongation cycle. Six

known metabolites (6–11) and complex fatty acid mixtures (FAMA-FAMG) iso-

lated from the Turkish marine sponge Agelas oroides were identified as active

components in the target (Pf FabI)-based antimalarial screening. Compounds 6–11showed inhibitory activity against Pf FabI with IC50 values of>100,>100, 40, 0.30,

5.0,>100, and>100 mg/mL, respectively. Interestingly, TFA salt of 8 showed betterinhibitory activity (IC50 40 mg/mL), while the activity of TFA salt of 9 became

weaker (IC50 5.0 mg/mL). These enzyme inhibitory activities were well correlated

with anti-plasmoidal activities against P. falciparum, except that 10 showed better

anti-protozoan activity (IC50 7.4 mg/mL) compared to its enzyme inhibitory activity.

1148 Y. Nakao and N. Fusetani

Page 5: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

Mixtures of fatty acids showed Pf FabI inhibitory activity with IC50s in the range of

0.35–20 mg/mL, while they showed anti-plasmoidal activities against P. falciparum(IC50s 3.4–40.9 mg/mL), Trypanosoma brucei rhodesiense (IC50s 5.3–42.3 mg/mL),

Trypanosoma cruzi (IC50s 16 to > 30 mg/mL), Leishmania donovani (IC50s

6.0–14.4 mg/mL), respectively. In addition, two mixtures (FAMA and FAMD)inhibited FabI of Mycobacterium tuberculosis (MtFabI, IC50s 9.4 and 8.2 mg/mL,

respectively) and Escherichia coli (EcFabI, IC50s 0.5 and 0.07 mg/mL, respectively).

FAMAwas a mixture of 5,9-dienoic fatty acids of C23(12) and C24(13), while FAMD

contained 10-methylhexadecanoic and palmitic acids as major lipids [4].

6

HOH

NH

Br

Br COOMe

7

NH

Br

Br COOH

8

NH

Br

Br

HN

ONH

N

NH2

9

H2NNH

N

NH2

10

+H3NS

O−

O

O

11

HO

O

n

12: n = 1113: n = 12

23.2.3 Lipoxygenases (EC 1.13.11.12)

Lipoxygenases (LO) mediate hydroperoxidation of polyunsaturated fatty acids

(PUFAS), leading to formation of leukotrienes and lipoxins. Selective inhibitors

of lipoxygenase isoforms can be useful for pharmacological agents, nutraceuticals,

23 Enzyme Inhibitors from Marine Invertebrates 1149

Page 6: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

or molecular tools. Fucosides, originally isolated from the Caribbean gorgonian

Eunicea fusca [5], selectively and irreversibly inhibited leukotriene (LT) formation

in murine models of inflammation. Further pharmacological study indicated that

fucoside B (14) inhibits conversion of arachidonic acid (AA) to leukotriene

B4 (LTB4) by inhibition of 5-LO with an IC50 of 18 mM [6]. Similarly,

didemnilactones A (15) and B (16) isolated from the tunicate Didemnum moseleyishowed inhibition against lipoxygenases of human polymorphonuclear leukocytes

with IC50 values of 9.4 and 8.5 mM (15 and 16, respectively against 5-LO), and

41 mM (15 against 15-LO) [7].

O

H H

O

OH

OH

OH

fucoside B (14)

O

O

OH

O

O

OH

didemnilactone A (15) didemnilactone B (16)

23.2.4 Indoleamine 2,3-Dioxygenases (EC 1.13.11.42)

Indoleamine-2,3-dioxygenase (IDO) catalyzes the oxidation of tryptophan to

N-formylkynurenine at the rate-limiting step in the metabolic degradation of trypto-

phan. Because T-cells are sensitive to tryptophan depletion, overexpression of IDO in

most tumors is thought to cause apoptosis of T-cells and, as the results, leads to immune

escape of solid tumors. Significant regression of tumors observed in a mouse model

system treated by a combination of relatively poor IDO inhibitor such as

1-methyltryptophan and cytotoxic agents (e.g., paclitaxel) have provided proof of

demonstration for the potential value of IDO inhibitors in cancer treatment.

Exiguamine A (17) was isolated as an IDO inhibitor from the marine sponge

Neopetrosia exigua collected at Papua New Guinea. The biogenesis of the unprec-

edented complex hexacyclic alkaloid skeleton of 17 was proposed to be starting

from DOPA, tryptophan, and N,N-dimethylhydantoin. Exiguamine A has a Ki of210 nM for inhibition of IDO in vitro [8]. Synthetic analogues of 17were preparedand evaluated their inhibitory activity against IDO. Although all of the synthetic

analogues turned out to be less active than 17 (Ki 41 nM in the modified

1150 Y. Nakao and N. Fusetani

Page 7: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

assay system), a simpler analogue 18 showed relatively potent noncompetitive

inhibition against IDO (Ki 200 nM) [9]. Four new polyketides annulin C (21),2-hydroxygarveatin, garveatin, and garvin C, as well as known annulins A (19) andB (20), garveatins A and C, and 2-hydroxy garvin A were isolated from the

Northeastern Pacific hydroid Garveia annulata. Among these compounds,

annulins A–C showed potent submicromolar IDO inhibition with Ki values of

0.14, 0.69, and 0.12 mM, respectively [10].

O

N

NH

O

O

H2N

OH

NN

O

O

exiguamine A (17)

NH

H2NO

O

NN

O

O

analogue (18)

O

O

OH

OO

OOR

annulin A (19) R = H

annulin C (21) R = Me

O

O

O

O

O

O OH

annulin B (20)

23.2.5 Neuronal Nitric Oxide Synthase (EC 1.14.13.39)

Neuronal nitric oxide synthase (nNOS) represents an important therapeutic target

for neuropathological disorders, such as stroke, Alzheimer’s disease, Parkinson’s

disease, AIDS, and dementia, because NO plays an important multifunctional role

in the modulation of many neurotransmitters in the brain and its overproduction has

been associated with such diseases. Eusynstyelamides A–C (22–24) isolated from

the Australian ascidian Eusynstyela latericius inhibited nNOS with IC50 values of

41.7, 4.3, and 5.8 mM, respectively, while they were found to be nontoxic toward

the three human tumor cell lines MCF-7 (breast), SF-268 (CNS), and H-460 (lung).

Eusynstyelamides A and B were mildly antibacterial against Staphylococcus aureus(IC50s 5.6 and 6.5 mM, respectively), and weakly inhibited the C4 plant regulatory

enzyme pyruvate phosphate dikinase (PPDK) (IC50 values of 19 and 20 mM,

respectively) [11].

23 Enzyme Inhibitors from Marine Invertebrates 1151

Page 8: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

eusynstyelamide A (22)

NH

Br

N

HN NH2

OOH

NH

HN

O

H OH

NH

Br

NH

NH2

NH

eusynstyelamide B (23)

NH

Br

N

HN NH2

OOH

NH

HN

O

H OH

NH

Br

NH

NH2

NH

eusynstyelamide C (24)

NH

Br

N

HN NH2

OOH

NH

HN

O

H OH

NH

Br

NH

NH2

NH

23.3 Transferases (EC 2.-.-.-)

23.3.1 1a-1,3-Fucosyltransferases (EC 2.4.1.214)

a-1,3-Fucosyltransferases (Fuc Ts, EC 2.4.1.214) which catalyze the transfer of

L-fucopyranoside residues from guanosine diphosphate fucose (GDP-fucose) to

glycoconjugate acceptors are known to be involved in the biosynthesis of sialyl

Lewis X (SLeX) present on the extracellular surfaces of leukocytes. The E-selectin-

SLeX interaction encourages leukocytes to move from the bloodstream into sites

of injury or infection and to cause inflammation, thus indicating that inhibitors

1152 Y. Nakao and N. Fusetani

Page 9: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

of a-1,3-fucosyltransferase are potential drugs for the treatment of inflammatory

diseases [12]. Octa- and nonaprenylhydroquinone sulfates 25 and 26 isolated from

an Australian marine sponge Sarcotragus sp. inhibited a-1,3-fucosyltransferase VII(Fuc TVII) with IC50 values of 3.9 and 2.4 mg/mL, respectively, while they showed

very weak activity against Fuc TVI [13].

OSO3Na

OH

n

octaprenylhydroquinone sulfate (25)

nonaprenylhydroquinone sulfate (26)

n6

7

23.3.2 Adenosine Phosphoribosyltransferase (EC 2.4.2.7)

Adenosine phosphoribosyltransferase (APRT) catalyzes the salvage pathway of

purine nucleotides. In mammals, there are both de novo and salvage pathways for

purine nucleotides synthesis, while kinetoplastid parasites such as Leishmaniaspp. lack the de novo pathway. Therefore, selective inhibitors of phosphoribosyl

transferase should compromise the parasite metabolism selectively. Three new

disulfated meroterpenoids, ilhabelanol (27), ilhabrene (28), and isoakaterpin (29)were isolated from a Brazilian sponge Callyspongia sp. as L. tarentolae adenosinephosphoribosyltransferase (L-APRT) inhibitors. Isoakaterpin (29) inhibited

L-APRT with an IC50 of 1.05 mM, while the accurate IC50 values for ilhabelanol

(27) and ilhabrene (28) could not be obtained because of the too small amount of

isolated compounds [14].

OSO3Na OSO3Na

NaO3SO NaO3SO

H

OH

H

ilhabelanol (27)

H

ilhabrene (28)

23 Enzyme Inhibitors from Marine Invertebrates 1153

Page 10: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

OSO3Na

NaO3SO

H

H

isoakaterpin (29)

23.3.3 Farnesyl Protein Transferase (EC 2.5.1.58)

The Ras family of guanine nucleotide binding proteins plays important roles

in signal transduction and regulation of cell differentiation and proliferation.

The Ras protein requires posttranslational processing in order to associate with

the plasma membrane and to function in signal transduction or cellular transfor-

mation. The first processing step is catalyzed by farnesyl protein transferase

(FPT) which adds a farnesyl group to a cystein residue near the carboxy terminus

of the Ras protein. Inhibition of FPT is a potential therapeutic target for novel

anticancer agents. A cembranolide diterpene 30 isolated from the soft coral

Lobophytum cristagalli showed potent inhibitory activity against FPT at an

IC50 of 0.15 mM [15]. The bioassay guided isolation of FPT inhibitors from the

marine sponge Hyrtios reticulate yielded a knowm sesterterpene, heteronemin

(31) as the active constituent which inhibited FPT with an IC50 value of 3 mM,

while its 12-epimer (32) did not show any noticeable activity [16].

Solandelactones C (33), D (34), and G (35) were cyclopropyl oxylipins isolated

from the hydroid Solanderia secunda and exhibited moderate inhibitory activity

against FPT (69, 89, and 61% inhibition at 100 mg/mL, respectively) [17].

Spongolactams A–C (36–38) were FPT inhibitors isolated from an Okinawan

marine sponge Spongia sp. by employing newly developed LC/MS-guided assay

systems. To study their structure–activities relationships, spongolactams were

semisynthesized from the known diterpene. The inhibitory activities of 36–38against FPT were confirmed as IC50 values of 23, 130, and >260 mM, respec-

tively [18]. Four new dual inhibitors, alisiaquinones A–C (39–41) and

alisiaquinol (42), against FPT and plasmoidal kinase Pfnek-1 were isolated

from an unidentified New Caledonian deepwater sponge. Compounds 39–42showed FPT inhibition at IC50 values of 2.8, 2.7, 1.9, and 4.7 mM, respectively.

1154 Y. Nakao and N. Fusetani

Page 11: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

Moreover, alisiaquinone A and alisiaquinol strongly inhibited plasmoidal NIMA-

like kinase Pfnek-1 at as low as 1 mM, whereas alisiaquinone C (41) showed

almost no inhibition at this concentration. However, alisiaquinone C showed

a much better inhibitory activity against P. falciparum in vitro. These results

correlated with a better activity of alisiaquinone C on FPT, as was shown by the

dramatic effects of FPT inhibitors on malaria parasites. The in vivo activity of

alisiaquinones A and C was investigated on rodent malaria and was shown that

they reduced by 50% the parasitemia at 5 mg/kg and displayed relatively high

level of toxicity with 100% and 80% mortality, respectively at 20 mg/kg, which

precluded further antimalarial development [19].

O

OAc

O

H

H O

OAcO

OAc

R

30heteronemin (31) R = β-OH

12-epi-heteronemin (32) R = α-OH

O

H

H OH

R1R2

OH

R1

OH

H

R2

H

OH

O

H

H OH

OH

OH

solandelactone C (33)

solandelactone D (34)

solandelactone G (35)

N

N

HN

COOH

X

Y

NCOOH

COOH

O

spongolactam A (36) X = H2, Y = O

spongolactam B (37) X = O, Y = H2

spongolactam C (38)

23 Enzyme Inhibitors from Marine Invertebrates 1155

Page 12: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

O

O

O

OOH

H

O

HO

OH

OOH

H

O

O

O

OOH

H

R SO2

HN

alisiaquinone A (39) R = H

alisiaquinone B (40) R = OMe

alisiaquinone C (41) alisiaquinol (42)

23.3.4 Geranylgeranyltransferase Type I (EC 2.5.1.59)

Geranylgeranyltransferase type I (GGTase I) catalyzes the posttranslational attach-

ment of the geranylgeranyl unit on the carboxy terminal cysteine residues of pro-

teins, to promote membrane interaction and biological activities of these proteins

[20]. Rho1p, a regulatory subunit of 1,3-b-D-glucan synthesis and the key player in

cell wall biosynthesis [21], is known as one of the targets of GGTase I and is

essential for viability of Saccharomyces cerevisiae. Since there is only 30%

sequence homology between the human and pathogenic fungus Candida albicansGGTase I [22], its inhibitors are expected to be selective antifungal agents.

Corticatic acids are the polyacetylenic GGTase I inhibitors isolated from the

marine sponge Petrosia corticata. Corticatic acids A (43), D (44), and E (45)inhibited C. albicans GGTase I with IC50 values of 1.9, 3.3, and 7.3 mM, respec-

tively, while corticatic acid A (43) also inhibited the growth of C. albicans with an

MIC value of 54 mM [23].

Massadine (46), a highly oxygenated alkaloid isolated from the marine sponge

Stylissa aff. massa as an inhibitor of GGTase I from C. albicans inhibited GGTase

I with an IC50 value of 3.9 mM [24].

OH

OH

O

Corticatic acid A (43)

OH

O

OHOH

Corticatic acid D (44)

1156 Y. Nakao and N. Fusetani

Page 13: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

OH

OOH

Corticatic acid E (45)

ONH

HN

HN

NH

NH

NH

NH2

H2NOH

O

O HN

HN

Br

BrBr

Br

OH

+

+

Massadine (46)

23.3.5 HIV Reverse Transcriptase (EC 2.7.7.49)

Reverse transcriptase (RT) is the key enzyme in the life cycle of human immuno-

deficiency virus (HIV). RT is a multifunctional enzyme responsible for the

transcription of viral RNA into double-stranded DNA, and exhibits both

RNA-dependent DNA polymerase (RDDP) and DNA-dependent DNA polymerase

(DDDP) activities as well as an inherent ribonuclease H (RNase H) activity. All the

catalytic functions of RT play a pivotal role in HIV replication. Taurospongin

A (47), a metabolite of an Okinawan sponge Hippospongia sp., is another example

of acetylenic compounds which possess HIV RT inhibitory activity (IC50 6.5 mM,

Ki 1.3 mM), along with inhibitory activity against DNA polymerase b (IC50 7.0 mM,

Ki 1.7 mM) and c-erbB-2 kinase (IC5028 mg/mL), but no cytotoxicity

(IC50>10 mg/mL) against L1210 and KB cells [25]. Tryptophan-derived pigments

48 and 49 and accompanying sesterterpenes 50 and 51 isolated from the Fijian

sponge Fascaplysinopsis reticulata showed weak inhibition against HIV RT [26].

Toxicols A–C (52–54) and toxiusol (55), triterpenes isolated from the Red Sea

sponge Toxiclona toxius, were reported to be inhibitory against HIV RT, but no

detailed bioactivities were available [27]. Clathsterol (56) isolated from a Red Sea

sponge Clathria sp. showed anti-HIV-1 RT at 10 mM [28]. Polycitone A (57),a general inhibitor of retroviral reverse transcriptases and cellular DNA polymer-

ases, was isolated from an ascidian Polycitor sp. It inhibited RDDP and DDDP

activity of HIV-1 RT with IC50 values of 245 and 470 nM, respectively, while it

showed general inhibition against MuLV (murine leukemia virus) RT, MMTV

(mouse mammary tumor virus) RT, calf-thymus pol a, human pol b, and KF

(E. coli DNA polymerase I) with IC50 values ranging from 73 to 600 nM [29].

23 Enzyme Inhibitors from Marine Invertebrates 1157

Page 14: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

HO3S

HN

O OH OAc O

O

OOR1

OR2R1

SO3Na

H

SO3Na

R2

SO3Na

H

H

taurospongin A (47)

NH

N

O

+Cl−

48

NH

N

OH

O

+

49

O

O

O

50

OH

OH

O

O51

toxicol A (52)

toxicol B (53)

toxicol C (54)

OSO3Na

NaO3SO

HH

toxiusol (55)

N

OH

Br

BrBrHO

BrBr

HO

Br

OH

Br

Br

OH

O O

O

OOH

OAc

NaO3SO

NaO3SO

O

O

clathsterol (56) polycitone A (57)

23.3.6 HIV Integrase (EC 2.7.7.-)

HIV integrase catalizes the initial DNA-breaking and DNA-joining reactions

responsible for the attachment of HIV cDNA to host DNA. Since there are no

similar proteins known to be important for normal function of cells, HIV integrase

1158 Y. Nakao and N. Fusetani

Page 15: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

is a promising target for less toxic anti-HIV drugs. A series of ascidian alkaloids,

the lamellarins (58)–(63) showed selective inhibition of HIV integrase with IC50

values of 16–73 mM for terminal cleavage and 14–51 mM for strand transfer

activities, respectively; lamellarin a 20-sulfate (58) showed inhibition in the early

steps of replication of HIV-1 in cell culture with an IC50 value of 8 mM [30].

Cyclodidemniserinol trisulfate (64) isolated from the Palauan ascidian Didemnumguttatum showed inhibition against HIV integrase at an IC50 of 60 mg/mL [31].

Haplosamates A (65) and B (66) isolated from two haplosclerid sponges

(Xestospongia sp. and unidentified sponge) inhibited HIV-1 integrase with IC50

values of 50 and 15 mg/mL, respectively [32]. Prenylhydroquinone sulfates 67 and

68 from a deepwater sponge Ircinia sp. showed HIV-1 integrase inhibition (65%

inhibition at 1 mg/mL and 45% inhibition at 5 mg/mL, respectively) [33].

O

N

RO O

MeO

HO

MeOYMeO

X

R

SO3Na

H

H

X

H

OMe

H

Y

OMe

OMe

OH

lamellarin α 20-sulfate

lamellarin W

lamellarin N

(58)

(59)

(60)

O

N

RO O

MeO

HO

MeOOMeMeO

X

Y R

SO3Na

SO3Na

H

X

H

OMe

OMe

Y

H

OH

H

NH

O

OO

O

O

OO

NaO3SO O

NHSO3Na

OSO3Na

cyclodidemniserinol trisulfate (64)

lamellarin U 20-sulfate

lamellarin V 20-sulfate

lamellarin T

(61)

(62)

(63)

23 Enzyme Inhibitors from Marine Invertebrates 1159

Page 16: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

O

NaO3SO

OH OH

OSO3NaOPO2HNa

haplosamate B (66)

68

H

OHOH

OH

6

O

H

H

H

OH

OHH

OH

NaO3SOO

P

O

OMeNaO

haplosamate A (65)

H

OH

OSO3Na

n

67: n = 5

23.3.7 Telomerase (EC 2.7.7.49)

Telomerase is a ribonucleoprotein enzyme that adds repeats of the DNA sequence,

TTAGGG, called telomere, onto the 3”-ends of chromosomes [34]. Telomerase

activity is found in about 90% of human tumors, but not in normal cells [35]. Thus,

inhibitors of telomerase are potential antitumor agents [36]. In fact, some synthetic

inhibitors based on the function of telomerase have been successful in clinical trials

[37]. From the marine sponge Dictyodendrilla verongiformis collected in southern

Japan, five telomerase inhibitors, dictyodendrins A–E (69–73) were isolated along

with the sodium salts of two known compounds (2 and 3) which were obtained as

aldose reductase inhibitors from a marine sponge of the same genus [3]. All these

seven compounds showed 100% inhibition of telomerase activity at the concentra-

tion of 50 mg/mL, while the desulfated analogue of dictyodendrin C (71) was notactive at this concentration [38]. Recently, the total synthesis of dictyodendrin

B (70), C (71), and E (73) was accomplished [39]. An unprecedented highly

sulfated lipopolysaccharide, axinelloside A (74), isolated from the marine sponge

Axinella infundibula showed a potent inhibition against human telomerase

(IC50 2.0 mg/mL) [40].

1160 Y. Nakao and N. Fusetani

Page 17: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

N

NH

OH

OH

OHMeO

O

HO

HO OSO3Na OSO3Na

OSO3Na

OSO3Na

OSO3Na

N

NH

OH

OH

OHO

HO

HO

N

NH

OH

OH

OO

HO

N

NH

OH

OO

HO

Dictyodendrin A (69) Dictyodendrin B (70)

Dictyodendrin C (71) Dictyodendrin D (72)

N

NH

OH

OH

O

HO

HO OSO3Na

Dictyodendrin E (73)

23 Enzyme Inhibitors from Marine Invertebrates 1161

Page 18: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

O

O

O

O

O

OO

OHXO

XO

MeXO

O

XO

OX

O

O

OH OH

O

OXO

Me

O

OXO

Me

OH

OO

XOO O

OH

XOO

O

MeO

XOO

XOOH

XOO

OMe

OH

O

XO

O

OH

OX

OXO

HOOH

OXO

O

OX

XO

OXHO

O

Axinelloside A (74)

X = SO3Na

23.3.8 Epidermal Growth Factor Receptor Kinase (EC 2.7.10.1)

Protein tyrosine kinases comprise a large family of enzymes that regulate cell

growth and intracellular signaling pathways; inhibitors of these enzymes may be

potential anticancer drugs [41]. There are at least four members of the type 1 growth

factor receptor gene family, including the epidermal growth factor receptors

(EGFR), erbB-1, erbB-2, erbB-3, and erbB-4. Members of this tyrosine kinase

receptor family have been implicated in the establishment or progression of

human cancer, particularly breast cancer, and enormous amounts of effort have

been made to identify specific small molecule inhibitors of EGFR tyrosine kinase

[42]. Tauroacidins A (75) and B (76), bromopyrrole metabolites of an Okinawan

spongeHymeniacidon sp., exhibited inhibitory activity against c-erbB-2 kinase withan IC50 of 20 mg/mL [43]. (+)-Aeroplysinin-1 (77) isolated from the marine sponge

Verongia aerophoba inhibited EGF receptor kinase completely at 0.5 mM [44].

Ma’edamine A (78), a cytotoxic bromotyrosine alkaloid isolated from a marine

sponge Suberea sp., inhibits c-erbB-2 kinase with an IC50 value of 6.7 mg/mL [45].

HN

OH

NH

NHHN

Br

R

O

NH

HNSO3

+

RBrH

OMe

Br Br

HOHO CN

tauroacidin A (75)tauroacidin B (76)

(+)-aeroplysinin-1 (77)

N

HNO

Br

MeO

O NMe

Br

Br Mema'edamine A (78)

1162 Y. Nakao and N. Fusetani

Page 19: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

23.3.9 Tyrosine Kinase pp60V-SRC (EC 2.7.10.2)

A tyrosine kinase, pp60V-SRC is the oncogenic protein encoded by Rous sarcoma virus.

Halenaquinone (79), halenaquinol (80), halenaquinol sulfate (81), and xestoquinone

(82) isolated from the Fijian sponge Xestospongia cf. carbonaria was found

to inhibit the kinase activity of pp60V-SRC with IC50 values of 1.5, 60, 0.55, and

28 mM, respectively [46]. Halenaquinone (79) also inhibited phosphatidylinositol

3-kinase with an IC50 value of 3 mM [47]. Two more pp60V-SRC inhibitors,

14-methoxyhalenaquinone (83) and xestoquinolide A (84) were isolated from the

same sponge (IC50 values of 5 and 80 mM, respectively) [48]. Melemeleone B (85),a sesquiterpene quinone isolated from twoDysidea sponges, showed inhibitory activityagainst pp60V-SRC with an IC50 value of 28 mM [49].

O

O O

O

O

NaO3SO

OH

O

O O

halenaquinol sulfate (81)halenaquinone (79)

HO

OH

O

O O

halenaquinol (80)

HO

OHN

SO3H

O

O O

O

O

OMe

O

O

O

O

melemeleone B (85)

14-methoxyhalenaquinone (83)

xestoquinolide B (84)

O

O O

O

O

xestoquinone (82)

23 Enzyme Inhibitors from Marine Invertebrates 1163

Page 20: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

23.3.10 Other Tyrosine Protein Kinase

Four prenylhydroquinone sulfates 67–68 and 90–91 obtained from a deepwater

sponge Ircinia sp. showed inhibitory activity of tyrosine phosphorylation by mem-

brane fraction prepared from HER2 overexpressing 3T3 cells with IC50 values of 8,

4, 8, and 5.9 mg/mL, respectively. Compounds 67 and 68 were also reported to be

HIV-1 integrase inhibitors [33].

23.3.11 MSK1 (EC 2.7.11.1) and MAPKAPK-2

Mitogen- and stress-activated kinase 1 (MSK1) and mitogen-activated protein

kinase–activated protein kinase 2 (MAPKAPK2) are two serine protein kinases

involved in signal transduction. Both of these enzymes are located in the nucleus

and are involved in the late stage of signal transduction pathway, therefore,

selective inhibitors of these enzymes are most likely to exhibit highly specific

cellular effects. Four cheilanthane sesterterpenoids 86–89 were isolated from

a marine sponge Ircinia sp. as inhibitors of both MSK1 (IC50s 4 mM for all

compounds) and MAPKAPK-2 (IC50 90 mM for all compounds) [50].

23.3.12 Raf/MEK1/MAPK

The Ras-MAPK-signaling cascade is found in all eukaryotic organisms and is

involved in cellular signaling processes. Since the oncogenic form of Ras is

associated with 30% of all cancers, Ras and the downstream kinases represent

attractive targets for pharmacological intervention. The Raf/MEK-1/MAPK cas-

cade inhibition assay-guided fractionation of the Philippine marine sponge Stylissamassa yielded eight known pyrrole alkaloids, aldisine (92), 2-bromoaldisine (93),10Z-debromohymenialdisine (94), a 1:1 mixture of 10E- and 10Z-hymenialdisines

(95–96), hymenin (97), oroidin (98), and 4,5-dibromopyrrole-2-carbonamide (99),among which 93–97 were active with IC50s ranging from 3 to 1288 nM; the most

active were 95 and 96 (IC50s 3 and 6 nM, respectively). In addition, all compounds

showed essentially identical IC50 values in the MEK-1 to MAPK assay, while none

of them showed activity in the Raf to MEK-1 assay [51].

23.3.13 Checkpoint Kinases (EC 2.7.11.1)

The synthetic10Z-debromohymenialdisine (94) inhibited checkpoint kinases

Chk1 and 2 with IC50 values of 3 and 3.5 mM, respectively [52]. Further investiga-

tion into its inhibitory activity toward kinases led to identification of 11 new targets

including p90RSK, KDR, c-Kit, Fes, MAPK1, PAK2, PDK1, PKCy, PKD2, Rsk1,and SGK for this class of alkaloids [53].

1164 Y. Nakao and N. Fusetani

Page 21: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

OHH

O

HO

HH

O

OH

O

H

86 87

HH

OO

OH

88

OHH

O

HO

OH

89

NHNH

O

O

R

R = HR = Br

aldisine (92)2-bromoaldisine (93)

NHNH

O

Br

NHN

O

NH2

10E-hymenialdisine (95)

NHNH

O

R

N

HN

H2N

hymenin (97)

H

OH

OSO3Na

n

90: n = 691: n = 7

NHNH

O

R

N

HN O

H2N

R = HR = Br

10Z-debromohymenialdisine (94)10Z-hymenialdisine (96)

HN

O

N

HN

NH

Br

Br

NH2

oroidin (98)

NH2

O

NH

Br

Br

99

23 Enzyme Inhibitors from Marine Invertebrates 1165

Page 22: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

23.3.14 Protein Kinase C (EC 2.7.11.13)

Protein kinase C (PKC), a phospholipid-dependent protein phosphorylating

enzyme, is a key player of cellular signal transduction and is believed to be

responsible for cancer, cardiovascular and renal disorders, and immunosuppres-

sion and autoimmune diseases, such as rheumatoid arthritis. Therefore, inhibi-

tors of protein kinase C (PKC) can be potential drug leads for the treatment of

such diseases. Staurosporine (100), isolated from Streptomyces staurosporeus, isthe most famous natural product PKC inhibitor [54]. 11-Hydroxystaurosporine

(101) isolated from a tunicate Eudistoma sp. collected in Pohnpei inhibited PKC

with an IC50 value of 2.2 nM which is about 30% more active than

staurosporine (100) [55]. Staurosporine aglycone K252-c (102) isolated from

another Eudistoma sample inhibited eight cloned PKC isoenzymes a, b I, b II,

d, e, Z, g, and z with IC50 values of 1.3, 0.6, 0.5, 1.2, 1.1, 0.8, 1.5, and

>6.4 mM, respectively [56]. Xestocyclamine A (103), bis-alkylpiperadinesfrom a marine sponge Xestospongia sp., moderately inhibited PKCe(IC50 4 mg/mL) [57]. Isoaaptamine (104), which was first reported from

a suberitid sponge in 1988, was found to be a PKC inhibitor; recent evaluation

of PKC inhibitory and antitumor activities for synthetic isoaaptamine and its

derivatives disclosed that none of them met the criteria for further evaluation

after the primary screen in the NCI tumor cell line panel [58]. Z-axinohydantoin(105) and debromo-Z-axinohydantoin (106) isolated from the marine sponge

Stylotella aurantium, moderately inhibited PKC (IC50s 9.0 and 22 mM, respec-

tively) [59].

HN

N N

O

RO

HN

OMe

MeRH

OH

HN

NH

NH

O

R

hydroxy staurosporine (100)

11-hydroxy staurosporine (101)

N

N

OH

xestocyclamine A (103)

K252-c (102)

1166 Y. Nakao and N. Fusetani

Page 23: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

N

OH

MeO N

NHNH

HN

HN

X

O

O O

XBr

H

isoaaptamine (104)

Z-axinohydantoin (105)

debromo-Z-axinohydantoin (106)

The mixture of corallidictyals A (107) and B (108), spirosesquiterpene alde-

hydes isolated from the marine sponge Aka (Siphonodictyon) coralliphagum,inhibited PKC with an IC50 value of 28 mM. Interestingly, the corallidictyal mixture

selectively inhibited PKCa with an IC50 value of 30 mM (IC50s 89, >300, and

>300 mM for e, Z, and z, respectively). In addition, the corallidictyal mixture

inhibited the growth of cultured Vero (African green monkey kidney) cells with an

IC50 value of 1 mM after continuous exposure (72 h) [60]. Nakijiquinones A–D

(109–112), sesquiterpene quinones isolated from an Okinawan marine sponge of

the family Spongiidae, exhibited inhibitory activity against PKC with IC50 values

of 270, 200, 23, and 220 mM, respectively, whereas they were also active against

EGF receptor kinases and c-erbB-2 kinase with IC50 values of >400, 250, 170, and

>400 mM, and 30, 95, 26, and 29 mM, respectively [61]. Recently, enantioselective

total synthesis of nakijiquinones and closely related analogues was reported [62].

The evaluation of their biological activities disclosed that C-2 epimer of

nakijiquinone (113) was a potent and selective inhibitor of tyrosine kinase

VEGFR2 (KDR) with an IC50 value of 21 mM. Incidentally, VEGFR2 is a receptor

for the vascular endothelial growth factor (VEGF) family and is responsible for

endothelial cell proliferation and blood vessel permeability, therefore inhibitors

of VEGFR2 are potential anti-angiogenic drugs [62]. Frondosines A–E (114–118),sesquiterpenoids from the sponge Dysidea frondosa, inhibited PKC with IC50

values of 1.8, 4.8, 20.9, 26, and 30.6 mM, respectively [63].

O

OHO

CHO

H

nakijiquinone A (109)nakijiquinone B (110)nakijiquinone C (111)nakijiquinone D (112)

2-epi-nakijiquinone (113)

corallidictyal B (108)

O

OHO

CHO

H

corallidictyal A (107)RGlyL-ValL-SerL-ThrL-Ser

H

O

O

HO

R

2

23 Enzyme Inhibitors from Marine Invertebrates 1167

Page 24: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

O

HO

frondosin B (115)

O

OH

frondosin C (116)

O

ORO

frondosin D (117)

frondosin E (118)

RH

Me

OH

HO

frondosin A (114)

Spongianolides A–E (119–123), cytotoxic sesterterpenes isolated from

a marine sponge Spongia sp., inhibited PKC at IC50 values of 20–30 mM [64].

Three secosterols 124–126 isolated from a gorgonian Pseudopterogorgia sp.

inhibited the human PKC a, bI, bII, g, d, e, Z, and z with IC50 values in the

range of 12–50 mM. The semisynthetic derivatives 127–129 also showed similar

activity [42].

O

O

O

O

OH

OH

H

H

OH

O

O

O

O

OH

OH

H

H

spongianolide A (119) spongianolide B (120)

H

H

OR

O

OH

OH

RCOCH3

COCH3

COCH(OH)CH3

spongianolide A (121) 16R

spongianolide B (122) 16S

spongianolide C (123) 16R

16

1168 Y. Nakao and N. Fusetani

Page 25: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

HOOC

H

O

HO

O128

HOOC

H

HOH

HO

OH129

HO

H

HO

HO

OH

126

HO

H

HO

HO127

HO

H

HO

HOH 125124

HO

H

HO

HOH

OH

Penazetidine A (130), an azetidine isolated from the Indo-Pacific sponge

Penares sollasi, inhibited PKC with an IC50 value of 1 mM [65]. BRS1 (131),a C30 bis-amino, bis-hydroxy polyunsaturated lipid from an unidentified Australian

sponge of the class Calcarea, inhibited not only PKC (EC50 98 mM) but also

radiolabeled phorbol ester binding to the enzyme (EC50 9.2 mM) [66]. Shimofuridin

A (132) isolated from an Okinawan marine tunicate Aplidium multiplicatuminhibited PKC at an IC50 of 20.0 mg/mL [67]. Bryostatin-1 (133) which was isolatedfrom the bryozoa Bugula neritina as an anticancer agent [68], is known to bind the

C1 domain of PKC and regulate its activity [69].

23 Enzyme Inhibitors from Marine Invertebrates 1169

Page 26: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

NH

HO

HO

NH2

OH

OH

NH2

N

NHN

OH

N

OHO

OHO

O

OH

O

O

O

O

O

BRS1 (131)

shimofuridin A (132)

penazetidine A (130)

O O

O

MeO

O O

O

O

O

OH

H

OMe

O

O OH

HO

O

bryostatin-1 (133)

23.3.15 Cyclin-Dependent Kinases (EC 2.7.11.22)

Cyclin-dependent kinases (CDK) are also a family of protein tyrosine kinases, some of

which are involved in cell cycles, and their inhibitors are expected to be potential

anticancer drugs. Konbu’acidin A (134) is a bromopyrrole alkaloid possessing inhib-

itory activity against CDK4 (IC50 20 mg/mL) isolated from a marine sponge

Hymeniacidon sp. [70]. Spongiacidins A (135) and B (136) also isolated from the

same sponge inhibited c-erbB-2 kinase (IC50s 8.5 and 6.0 mg/mL, respectively) and

cyclin-dependent kinase 4 (CDK4) (IC50s 32 and 12 mg/mL, respectively) [71].

Rhopaladins A–D isolated from an Okinawan tunicate Rhopalaea sp. are the first

example of bis-indole alkaloids possessing an imidazolinone moiety as tunicate

metabolites; rhopaladin B (137) inhibited CDK4 and c-erbB-2 kinasewith IC50 values

of 12.5 and 7.4 mg/mL, respectively [72]. Microxine (138) from an Australian marine

spongeMicroxina sp. inhibited CDK1 (cdc2) kinasewith an IC50 value of 13 mM[73].

NHN

HNHN

NH

H2N

Cl

NH

O HN

H2N

Br

OHO

Br

Br

H

H

+

+

NHNH

NHHN

NH2

O

O

Br

R

+

konbu'acidin (134) spongiacidin A (135) R = Br

spongiacidin B (136) R = H

1170 Y. Nakao and N. Fusetani

Page 27: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

NH

N

NH

NH

O

HO

O

N

N

NH

MeN

HN

SO3H

O

Rhopaladin B (137) microxine (138)

23.3.16 Glycogen Synthase Kinase-3 (2.7.11.17)

Glycogen synthase kinase-3 (GSK-3) is a serine-threonine kinase ubiquitously

expressed and involved in the regulation of many cell functions. GSK-3 phos-

phorylates glycogen synthase or the microtuble-associated protein tau, being

implicated in type-2 diabetes, Alzheimer’s disease, or different tau pathologies

such as Pick’s disease. Hymenialdisine (96) [74], indirubine (139) [75], and

meridianine (140) [76] are the known marine-derived GSK-3 inhibitors isolated

from several species of marine sponges, the mollusk Hexaplex trunculus, and the

ascidian Aplidium meridianum, respectively. Hymenialdisine (96) potently

inhibited GSK-3b (IC50 10 nM) and plant GSK-3 (ASK�g) with an IC50 value

of 80 nM as well. It inhibited in vivo phosphorylation of specific neuronal

proteins by GSK-3b and CDK5 such as AD-characteristic phosphorylation of

tau completely [74]. Indirubin inhibited GSK-3a/b with an IC50 value of 1.00 mM[75]. Studies of structure–activity relationship (SAR) in the class of manzamine

alkaloids revealed that manzamine A (141) inhibited GSK-3b at an IC50 value of

10.2 mM; 8-hydroxymanzamine A (142) being the most potent analogue

(IC50 4.8 mM) [77].

NNH

NH

I

O

NHO

139

NH

NN

H2N

OH

Br140

N

NNH

OH

R

141: R = H142: R = OH

23 Enzyme Inhibitors from Marine Invertebrates 1171

Page 28: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

23.4 Hydrolases (EC 3.-.-.-)

23.4.1 Phospholipase A2 (EC 3.1.1.4)

Phospholipase A2 (PLA2) cleaves the ester linkage of the b-position of phospho-

lipids to release arachidonic acid which is metabolized to prostaglandins, leukotri-

enes, and other mediators of inflammation. Prostaglandins are implicated in various

forms of pain and inflammation. In biogenesis of prostaglandins, arachidonic acid

formation is the rate-determining step, thus, PLA2 inhibitors are expected to be

potential anti-inflammatory drugs. Manoalide (143), initially isolated as an

antibacterial metabolite from the marine sponge Luffariella variabilis [78], was

later found to be a potent inhibitor of PLA2 with an IC50 value of 1.7 mM [79], which

led to synthesis and anti-inflammatory evaluation of more than 100 analogues.

Unfortunately, none of them was developed for drugs. Three congeners of

manoalide were also obtained from the same sponge [80], of which secomanoalide

(144) is more potent against bovine pancreatic PLA2 [81]. Luffariellolide (145)isolated from a Palauan sponge Luffariella sp. also inhibited the bee venom PLA2

with an IC50 of 0.23 mM. The partially reversible and weaker inhibitory activity of

145was suggesting that the g-lactol group in 143was responsible for the irreversiblereaction of 143 with Lys residues on PLA2 [82]. Luffariellins A (146) and B (147)isolated from L. variabilis are very potent against bee venom PLA2 with IC50 values

of 56 and 62 nM, respectively [83]. These sesterterpenes are known to covalently

and specifically modify PLA2 by a Schiff base formation between Lys-56 residue of

PLA2 and the hemiacetal or aldehyde functionality of these terpenoids [84].

According to this mechanism, Katsumura and coworkers designed PLA2 inhibitors,

one of which potently and selectively inhibited bovine pancreas PLA2 [85].

O

OH

O

OH

HO

O

OH

O

OH

HO

O

OH

O

OHO

O

OH

OOHO

O

OH

Oluffariellolide (145)

manoalide (143)

secomanoalide (144)

luffariellin A (146)

luffariellin B (147)

1172 Y. Nakao and N. Fusetani

Page 29: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

Cacospongionolides (148–150), originally isolated as antitumor compounds

from the Mediterranean sponge Fasciospongia cavernosa [86], preferentially

inhibited bee venom and human synovial PLA2 among PLA2s of various origins.

Cacospongiolide B (149) exhibited specific inhibition against human PLA2 [87],

while cacospongionolide E (150) was the most potent inhibitor toward human

synovial PLA2 (IC50 1.4 mM), showing higher potency than manoalide [88]. The

structure–activity relationship study on synthetic analogues suggested that 149 has

an enantiospecific interaction with the enzyme that is independent of the

g-hydroxybutenolide moiety [89]. Furthermore, cacospongionolide B (149) was

reported to suppresses the expression of inflammatory enzymes including

inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) as well as

to reduce tumor necrosis factor-a (TNF-a) [90]. Cavernolide (151) isolated from

a marine sponge Fasciospongia cavernosa selectively inhibited human PLA2 with

an IC50 value of 8.8 mM [91].

O

OH

HO

OH

HO

O

cacospongionolide B (149)cacospongionolide (148)

OO

O

OH

H OH

O

cacospongionolide E (150) cavernolide (151)

Dysidiotronic acid (152), a sesquiterpenoid from a Vanuatu sponge Dysidea sp.,inhibited human synovial PLA2 with an IC50 value of 2.6 mM [92], while

bolinaquinone (153), dysidine (154), and a 1:1 mixture of dysideonones A (155)and B (156) significantly inhibited human synovial PLA2, among which 153showed the most potent activity with an IC50 value of 0.2 mM, but was not

a selective inhibitor toward this enzyme. On the contrary, dysidine (150) showedselective inhibition against this enzyme with an IC50 value of 2.0 mM. However,

153–156 did not inhibit cPLA2 [93].

H

O

O

O

OHMeO

H

O

O

OH

OH

dysidiotronic acid (152) bolinaquinone (153)

23 Enzyme Inhibitors from Marine Invertebrates 1173

Page 30: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

H

H

HO

O

OMe

OOMe

dysidenone A (155)

H

H

HO

O

OMe

OOMe

dysidenone B (156)

H

O

O

OH

OH

NH

SO3Hdysidine (154)

Petrosaspongiolides M–R (157–161), sesterterpens from the New Caledonian

marine sponge Petrosaspongia nigra inhibited PLA2; the most potent petrosas-

pongiolide M (157) inhibited human synovial and bee venom PLA2 with IC50

values of 1.6 and 0.6 mM, respectively, while petrosaspongiolide P (159)inhibited human synovial PLA2 (IC50 3.8 mM) more selectively than bee venom

PLA2 (IC50 >10 mM). Other petrosaspongiolides N (158), Q (160), and R (161)showed only weak activity [94]. Petrosaspongiolide M was shown to form the

covalent adduct between PLA2 and g-hydroxybutenolide which is essential for the

activity [95]. Ironically, a mild non-covalent PLA2 inhibitor, 25-acetyl-

petrosaspongiolide M, was found to be hydrolyzed with PLA2 to form the potent

inhibitor, petrosaspongiolide M [96]. Interestingly, a full characterization of the bvPLA2 adduct with petrosaspongiolide R (161), one of the least active and most

structurally different among petrosaspongiolides, using LC-MS, MSn, and compu-

tational methods, confirmed the same inhibition mechanism and covalent binding

site already found for petrosaspongiolide M (167). The correct arrangement of the

ligand in the active site, induced by non-covalent interactions, is of pivotal

importance in the inactivation process [97]. The anti-inflammatory effects of

petrosaspongiolide M was suggested to be mediated by inhibition of NF-kBsignaling pathway [98].

Aplyolide (162), a sesterterpene isolated from the marine sponge

Aplysinopsis elegans inhibited human PLA2 with an IC50 value of 10.5 mM[99], whereas the related terpenoid, luffolide (163) isolated from a Palauan

Luffariella sp., completely inhibited bee venom PLA2 at a concentration of

3.5 mM [100].

1174 Y. Nakao and N. Fusetani

Page 31: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

R

O

HO

O

O

OAc

HH

H

H

O

OH

HH

H

H

R

O

HO

O

RHOAc

RHOAc

petrosaspongiolide M (157)N (158)

petrosaspongiolide P (159)Q (160)

O

H

H

O

HO

O

OH

H

H

O

HO

O

OH

petrosaspongiolide R (161) aplyolide A (162)

CHO

H

H

O

HO

O

OAc

luffolide (163)

A homoscalarane sesterterpene (164) isolated from a marine sponge

Lendenfeldia frondosa showed moderate PLA2 inhibition (35% inhibition at

8 mM) [101]. A sesterterpene, 12-deacetyl-23-acetoxy-20-methyl-12-epi-

scalaradial (165), from the Pacific nudibranchs Glossodoris sedan and G. dalliinhibited mammalian cytosolic PLA2 with an IC50 value of 18.0 mM [102]. The

parent compound, scalaradial (166) is more potent (IC50 0.6 mM) [84a, 103].

HH

H

AcO

OAc

OOO

AcO

OH CHO O

164 165

23 Enzyme Inhibitors from Marine Invertebrates 1175

Page 32: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

OAc

CHO

CHO

scalaradial (166)

Halisulfate 1 (167) from a halichondrid sponge inhibited PLA2 completely at

16 mg/mL, while a mixture of halisulfates 2–4 (168–170) inhibited PMA-induced

inflammation in the mouse ear edema assay as well as PLA2(data not shown) [104].

Palinurin (171), originally isolated from the Mediterranean marine sponge Irciniavariabilis [105], was inhibitory against PLA2 with an IC50 value of 50 mM [2]. Two

sesterterpenes, palauolol (172) and palauolide (173) isolated from a Palauan sponge

Fascaplysinopsis sp. inhibit bee venom PLA2 (85% and 82% inhibition at

0.8 mg/mL, respectively) [106].

OSO3Na

OH

HO

OSO3NaO

halisulfate-1 (167) halisulfate-2 (168)

NaO3SOO

halisulfate-3 (169)

NaO3SOO

halisulfate-4 (170)

OO

HO

palauolide (173)

O

OHO

O

palinurin (171)

OH

O

HO

O

palaulol (172)

1176 Y. Nakao and N. Fusetani

Page 33: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

Two polyhydroxy sterols 174 and 175 from the Korean gorgonian Acabariaundulata inhibited PLA2 with IC50 values of 13.8 and 21.5 mM, respectively [107].

Valdivones A (176) and B (177), diterpenoids isolated from the South African soft

coral Alcyonium valdivae, strongly inhibited chemically induced inflammation in the

mouse ear assay (93% and 72% inhibition at 50 mg/ear, respectively), but their activityagainst bee venom PLA2 was not potent (43% inhibition at 16 mg/mL for both) [108].

O

OH

O

O

O

O

OH

O

O

O

valvidone A (176) valvidone B (177)

HOOH

OH

O

HOOH

OH

O

174 175

23.4.2 Acetylcholinesterase (EC 3.1.1.7)

3-Alkylpyridinium polymers 178 from a marine sponge Reniera sarai showedpotent inhibitory activity against electric eel acetylcholinesterase (AchE), human

erythrocyte AchE, insect recombinant AchE, and horse serum butylcholinesterase

(BuChE) with IC50 values of 0.06, 0.08, 0.57, and 0.14 mg/mL, respectively, while it

did not inhibit trypsin or alkaline phosphatase [109].

N+

n

MW 5520 & 18900

178

23 Enzyme Inhibitors from Marine Invertebrates 1177

Page 34: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

23.4.3 Protein Phosphatases (EC 3.1.3.16)

Reversible protein serine/threonine phosphorylation regulates numerous cellular

functions, including muscle contraction, neurotransmission, cell proliferation, car-

cinogenesis, and apoptosis. Although a large number of protein kinases have been

well investigated for several decades, it is quite recent that our understanding of

protein phosphatases (PPs), namely PP1, PP2A, PP2B, and PP2C, has been con-

siderably deepened by the discovery of natural product inhibitors.

23.4.3.1 PP1 and PP2Okadaic acid (179), the first natural product inhibitor of PP1 and PP2A, is

a polyether polyketide isolated as a cytotoxic substance from two sponges, Japanese

Halichondria okadai and Caribbean H. melanodocia [110]. It was succeedingly

identified as a causative agent of the diarrhetic shellfish poisoning. Later, 179 was

found to show a potent tumor promoting activity caused by potent inhibition of PP1

and PP2A with IC50 values of 3 and 0.2–1 nM, respectively, whereas it showed only

weak or no inhibition against PP2B or PP2C [111]. It has become a powerful tool for

the study of biological processes mediated by protein phosphorylation. The crystal

structure of PP1g-okadaic acid complex was elucidated in 1995 [112]. A remarkable

reduction of inhibitory activity against PP1 (IC50 465 nM) was observed for 19-epi-okadaic acid isolated from dinoflagellate Prorocentrum belizeanum, whereas the

inhibitory activity against PP2A remained as the same level (IC50 0.47 nM) for 179.It was suggested that the stereochemistry at C19 is crucial for the selectivity between

PP1 and PP2A [113]. A new okadaic acid class compound, belizeanic acid (180)with an unusual skeleton was isolated from the dinoflagellate Prorocentrumbelizeanum. Despite its opening of C/D rings, it still retains the potent inhibitory

activity against PP1 (IC50 318 nM), slightly reduced potency compared to okadaic

acid (IC50 62 nM in the same assay system) [114].

HOO

O

OHO

OH

O

O

O O

O

HOH

H HOH

H

okadaic acid (179)

HOO

O

OHOH

O O

OH

OH

HO

OH

OH

O

O

(180)

19

1178 Y. Nakao and N. Fusetani

Page 35: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

Calyculin A (181) was isolated from the Japanese marine sponge Discodermiacalyx as a potent antitumor agent [115]. Like okadaic acid, it was found to be

a potent tumor promoter and to inhibit PP1 and PP2A with IC50 values of 1.4 and

2.6 nM, respectively, while it showed weak inhibition against PP2B [116].

Calyculins B–H (182–188), geometrical isomers of 181, or their 32-methyl deriv-

atives which were succeedingly isolated from the same sponge, were originally

reported to have similar inhibitory activity against PP2A (IC501.0–6.0 nM) [117],

but this was later found to be not correct; the geometry of the tetraene portion

affects the enzyme inhibition significantly [119]. Furthermore, calyculin J (189),calyculinamides A (190) and F (191), des-N-methylcalyculin A (192) and

dephosphonocalyculin A (193) were isolated as PP2A inhibitors from D. calyx[118]. Later, isolation of hemicalyculin A (194) from D. calyx and structure-

activity relationship studies were reported [119]. Hemicalyculin A (194) was

equipotent as calyculin A (181) against PP1g and PP2A with IC50 values of 14

and 1.0 nM, respectively. Interestingly, 194 showed cytotoxicity over 2000 times

less than that of 181. More recently, the crystal structure of the calyculin A-PP1gcomplex was solved by X-ray crystallography [120], which showed that the

hydrophobic tail, phosphate, and 13-OH moieties are essential for binding to

PP1g. These features are the cases of the PP1c/microcystin LR [121] and PP1c/

okadaic acid complexes [112]. Clavosines A–C (195–197), glycosylated deriva-

tives of C21-epi-calyculinamide C, were isolated from the marine sponge

Myriastra clavosa. Clavosines A (195) and B (197) inhibited PP1cg (IC50s 0.5

and 13 nM, respectively), native PP1c (IC50s 0.25 and 1.0 nM, respectively), and

PP2Ac (IC50s 0.6 and 1.2 nM, respectively) [122].

R1

CNHCNHCONH

R2

HCNHCNH

R3

HHMeMeH

calyculin A (181)calyculin B (182)calyculin C (183)calyculin D (184)

calyculinamide A (190)

MeO NH

Me2N

OH

OH

O

N

O

O

OHO

O

OMeOHOH

R1

R3

R2

PHO

O

HO

23 Enzyme Inhibitors from Marine Invertebrates 1179

Page 36: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

HR2

OH

OH

O

N

O

O

OHO

R1

OMeOHOH

CN

R1

OPO3H2

OH

R2

NHMe

NMe2

MeO N

des-N-methylcalyculin A (192)

dephosphonocalyculin A (193)

HMe2N

OH

OH

O

N

O

O

OHO

O

OMeOHOH

R3

R2

R1 PHO

O

HO

MeO N

R1

CN

H

CN

H

H

R2

H

CN

H

CN

CONH

R3

H

H

Me

Me

H

calyculin E (185)

calyculin F (186)

calyculin G (187)

calyculin H (188)

calyculinamide F (191)

MeO NH

Me2N

OH

OH

O

N

O

O

OHO

O

OMeOH

O

CNBr

PHO

O

HO

calyculin J (189)

1180 Y. Nakao and N. Fusetani

Page 37: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

MeO NH

Me2N

OH

OH

O

N

O

O

OO

O

OMeOHOH

R1

R2

PHO

O

HO

OMe

OMeOMe

MeOR1

CONH2

H6Z isomer of 196

R2

HCONH2

clavosine A (195)clavosine B (196)clavosine C (197)

6

NC

O

OHO

O

OMeOHOH

CN

PHO

O

HO

hemicalyculin A (194)

Motuporin A (198), one of the most potent PP1 inhibitors known, isolated from

the Papua New Guinea sponge Theonella swinhoei inhibited PP1 at <1 nM [123].

Dragmacidins D (199) and E (200) isolated from a southern Australian deepwater

sponge Spongosorites sp. are potent serine-threonine protein phosphatase inhibi-

tors; 199 selectively inhibited PP1, while 200 inhibited both PP1 and PP2A in the

preliminary test (data not shown). Dragmacidin E (200) seemed to exist in tauto-

meric equilibrium between pyrazine and pyrazinone forms [124]. Spirastrellolide

A (201) was a novel macrolide isolated from the marine sponge Spirastrellacoccinea as an antimitotic agent [125]. Recently, its revised structure and selective

inhibitory activity against PP2A (IC50 1 nM) were reported [126]. Nagelamides are

dimeric bromopyrrole alkaloids isolated from a marine sponge Agelas sp., among

which nagelamides A–C (202–204), G (205), and H (206) were shown to inhibit

PP2A (IC50 s 13 to >50 mM) [127].

23 Enzyme Inhibitors from Marine Invertebrates 1181

Page 38: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

NO

O

O

NH

O NH

OHO

OH

OHN

NHOMe O

N

HN

HN

NH

O

Br

OH

NH

HN

HN

motuporin A (198)

+

TFA−

dragmacidin D (199)

HO

O

OHO

OO

OO

HO

HO

OMeOH

HO

Cl

OMe

O

O

O

spirastrellolide A (201)

N

HN

HN

HN

NHNH

O

Br

H2N

OH

+

N

N

HN

HN

NHNH

HO

Br

H2N

OH

dragmacidin E (200)

+

HN

NH

HNNH

NH

NH

NH2

O

O

HN

NH

Br

Br

Br

Br

NH2

nagelamide C (204)

HN

NH

HNNH

NH

NH

NH2

O

O

HN

NH

Br

Br

Br

Br

NH2

R

+

+

nagelamide A (202)

B (203)

R = H

R = OH

1182 Y. Nakao and N. Fusetani

Page 39: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

NH

HN

NH2

NH

HN

H2N

HN

O

HN

O

HN

NH

Br

Br

Br

Br

+

+

nagelamide G (205)

NH

HNNH

NH2

OHN

Br

Br

NH

HN

HN

NH

Br

Br

O

NH

HN

SO3−

+

+

nagelamide H (206)

23.4.3.2 Calcineurin (PP2B)Calcineurin (CaN), a serine-threonine protein phosphatase (PP2B) involved in signal

transduction, is recognized as one of the principal signaling molecules that regulates

the immune response [128]. Immunosuppressants such as FK506 and cyclosporin

A have been shown to exert their effect through inhibition of CaN following their

association with binding proteins [129]. Thus, inhibitors of CaN may be useful as

immunosuppressants. Secobatzellines A (207) and B (208) isolated from a Caribbean

deepwater spongeBatzella sp. inhibitedCaNwith IC50 values of 0.55 and 2.21mg/mL,

respectively. Secobatzelline A (207) also inhibited peptidase activity of CPP32

(IC50 0.02 mg/mL), a cysteine protease known as caspase-3 which play a major role

in the programmed cell death known as apoptosis [130].

HN

O

R

Cl

H2N

OH

OH

secobatzelline A (207) R = NHsecobatzelline B (208) R = O

23.4.4 Protein Tyrosine Phosphatases (EC 3.1.3.48)

Cdc25 is a dual-specificity protein tyrosine phosphatase which regulates activa-

tion of CDKs (cyclin-dependent kinases) through the dephosphorylations of

a threonine and a tyrosine residues of CDKs. Due to their unique substrate

selectivity and their essential functions in cell cycle control, cdc25 phosphatases

are attractive screening target to identify new antimitotic compounds.

Dysidiolide (209) is the first natural product inhibitor of cdc25A isolated from

the Caribbean sponge Dysidea etheria. It inhibited the dephosphorylation of

p-nitrophenol phosphate by cdc25A phosphatase with an IC50 of 9.4 mM [131].

23 Enzyme Inhibitors from Marine Invertebrates 1183

Page 40: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

Because of its unusual structure and interesting biological activity, a number of

synthetic chemists entered the race of total synthesis of dysidiolide, from

which some important structure-activity relationships were provided [132].

Coscinosulfate (210), a sesquiterpene sulfate isolated from the New Caledonian

sponge Coscinoderma mathewsi exhibited selective inhibition against cdc25A

phosphatase with an IC50 of 3 mM [133].

O

O

OH

H

OSO3Na

HO

OH

dysidiolide (209) coscinosulfate (210)

23.4.5 Phospholipase C (EC 3.1.4.3)

Phosphatidylinositol-specific phospholipase C (PI-PLC) is the rate-limiting enzyme

in the phosphatidylinositol turnover. PI-PLC hydrolyzes the phosphate-glycerol

linkage of phosphatidylinositol to produce two second messengers, inositol triphos-

phate and diacylglycerol which play key roles in cellular signal transduction

induced by growth factors and hormones [134].

Akaterpin (211) isolated from an Okinawan marine sponge Callyspongia sp.

showed potent inhibition against PI-PLC (IC50 0.5 mg/mL) as well as weak inhibi-

tion against neutral sphingomyelinase with an IC50 value of 30 mg/mL [135].

NaO3SO

OSO3NaH

H

akaterpin (211)

1184 Y. Nakao and N. Fusetani

Page 41: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

23.4.6 Phosphodiesterase (EC 3.1.4.-)

The Ca2+-calmodulin system plays an important role in the control of cellular

proliferation and tumor formation. A calmodulin antagonist, W-7, has been found

to inhibit proliferation of Chinese hamster cells and formation of mouse skin tumors.

Eudistomin A (212), a b-carboline alkaloid isolated from the Okinawan tunicate

Eudistoma glaucus, inhibited calmodulin-activated brain phosphodiesterase with an

IC50 value of 20 mM [136], while eudistomin C (213) exhibited calmodulin antag-

onistic activity (IC50 30 mM) [137]. Pseudodistomins A (214) and B (215), unsualalkylpiperazines of the Okinawan tunicate Pseudodistoma kanoko, inhibited cal-

modulin-activated brain phosphodiesterase at the same concentration (IC50 30 mM),

which is about three times more potent than W-7 [138]. Rigidin (216) isolated froman Okinawan tunicate Eudistoma cf. rigida inhibited calmodulin-activated brain

phosphodiesterase with an IC50 value of 50 mM [139]. Stellettamide A (217),an unusual indolizidine alkaloid originally isolated from a marine sponge

Stelletta sp. as an antifungal and cytotoxic compound [140], was found to inhibit

Ca2+/calmodulin-dependent phosphodiesterase and (Ca2+-Mg2+)-ATPase with IC50

values of 52 and 100 mM, respectively [141]. Related alkaloids, stellettazole A (218)[142], bistellettadines A (219), and B (220) [143], from the same sponge showed

moderate inhibitory activity against Ca2+/calmodulin-dependent phosphodiesterase

(218: 45% inhibition at 100 mM, 219 and 220: 40% inhibition at 100 mM).

NH

HN

NH

O

O

O

OH

OH

NH

N

NOH

Br

NH

N

HO

SMeHN

Br

Me

NH

HO

HO

H2N

H2NNH

pseudodistomin A (214)

pseudodistomin B (215) rigidin (216)

eudistomin A (212) eudistomin C (213)

23 Enzyme Inhibitors from Marine Invertebrates 1185

Page 42: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

NH

NH

HN NH2

NH2

NH2

NH2

O

HN

HN

NH

H

O NH2

NH2

+

++

+

4X−

bistellettadine A (219)

NH

NH

HN NH2

O

HN

HN

NH

NH2

NH2

NH2

O NH2

NH2

+

++

+

4X−

220

HN

O

N

H

Me+ X−

HN

O N

N NH

NH2

NH2

Me

+

+

stellettamide A (217)

2X−

stellettazole A (218)

23.4.7 Sialidase (EC 3.2.1.18)

Sialidase cleaves an a-linked terminal N-acetylneuraminic acid from glycoproteins,

glycolipids, and oligosaccharides. In several viral and bacterial infections, neur-

aminidases play important roles; influenza virus employs this enzyme to detach

itself from the infected cell in the budding stage, thus indicating requirement of

neuraminidase for replication of the virus. Therefore, selective inhibitors

1186 Y. Nakao and N. Fusetani

Page 43: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

of sialidase are potential therapeutic agents against ifluenza. The a-glucosidaseinhibitors, schulzeines A–C (221–223), were also inhibitory against viral neuramin-

idase with IC50 values of 60 mM. Calyceramides A–C (224–226), sulfated

ceramides isolated from the marine sponge Discodermia calyx, inhibited sialidase

from Clostridium perfringens with IC50 values of 0.4, 0.2, and 0.8 mg/mL,

respectively [144]. Another example of a marine sialidase inhibitor is nobiloside

(227), a triterpenoid saponin of the marine sponge Erylus nobilis, which inhibitedClostridium perfringens sialidase with an IC50 value of 0.46 mg/mL [145].

Asteropine A (228) is the first cystine knot of sponge origin isolated from

Asteropus simplex. Asteropine A showed potent and competitive inhibition

against bacterial sialidases (from Clostridium perfringens, Vibrio choleae,and Salmonella typhimurium) with Ki values of 36.7, 340, and 350 nM, respec-

tively [146].

N

NH

OH

HO

OO

OSO3Na

NaO3SO

OSO3Na

schulzeine A (221)

N

NH

OH

HO

OO NaO3SO

OSO3Na

OSO3Naschulzeine B (222)

N

NH

OH

HO

OO

OSO3Na

NaO3SO

OSO3Na

schulzeine C (223)

23 Enzyme Inhibitors from Marine Invertebrates 1187

Page 44: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

HN

NaO3SO

OH

O

HO

calyceramide C (226)

COOH

OO

HOOC

OH

OO

OO

OH

COOHHO

OHHO

OH

HO

nobiloside (227)

YCGLFGDLCTLDGTLACCIALELECIPLNDFVGICL

asteropine A (228)

HN

NaO3SO

OH

O

HO

calyceramide A (224)

HN

NaO3SO

OH

O

HO

calyceramide B (225)

23.4.8 Chitinase (EC 3.2.1.14)

Chitinase plays crucial roles in a wide variety of organisms ranging from nutrition

to defence and control of ecdysis in arthropods, thus indicating their potential for

antifungal and insecticidal agents. Styloguanidines (229–231) isolated from the

1188 Y. Nakao and N. Fusetani

Page 45: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

sponge Stylotella aurantium collected off Yap showed inhibitory activity against

chitinase from a bacterium Schwanella sp. at the dose of 2.5 mg/disc by using the

“squid chitin agar plate method.”[147].

N

N

NH

NH

HN

N

H2N

O

H2N

HO

Cl NH2

R1R2

R1 R2

H HBr HBr Br

styloguanidine (229)3-bromostyloguanidine (230)2,3-dibromostyloguanidine (231)

23.4.9 a-Glucosidases (EC 3.2.1.20)

a-Glucosidases are involved in glycoprotein processing and glycogenolysis and,

therefore, their inhibitors can be applied for treatment of diabetes, obesity, viral

infections, and cancer. Penarolide sulfates A1(232) and A2(233), L-proline

containing macrolide trisulfates isolated from a marine sponge Penares sp.,

inhibited a-glucosidase of yeast origin with IC50 values of 1.2 and 1.5 mg/mL,

respectively, while they showed only weak or no inhibition against b-glucosidaseor b-galactosidase. Penarolide sulfates also inhibited thrombin with IC50 values

of 3.7–4.2 mg/mL. Perhaps, the three sulfate groups are responsible for the

activity [148]. A linear type congener, penasulfate A (234), in which L-proline

was replaced by D-pipecolic acid, was isolated from the same sponge.

Penasulfate A (234) showed about ten times as potent inhibition (IC50 0.14 mg/mL) as penarolides againt the same enzyme, while the inhibitory activity against

the enzyme from the different source remained the same level [149]. The known

1,4-dideoxy-1,4-imino-D-arabinitol (235) was isolated as an a-glucosidase inhib-itor from a marine sponge Haliclona sp. collected in Western Australia.

Iminopentitol (235) strongly inhibited yeast a-glucosidase at an IC50 value of

0.16 mg/mL [150]. Callyspongynic acid (236), a polyacetylenic acid was isolated

from the Japanese marine sponge Callyspongia truncata Callyspongynic acid (236)inhibited a-glucosidase with an IC50 value of 0.25 mg/mL, but was inactive against

b-glucosidase, b-galactosidase, thrombin, and trypsin at 100 mg/mL. Both the carbox-

ylic acid and the allylic alcohol linked to an acetylene are thought to be important for

the activity [151]. The Japanese marine sponge Penares schulzei afforded three new

a-glucosidase inhibitors, schulzeines A–C (221–223), which showed potent inhibitory

23 Enzyme Inhibitors from Marine Invertebrates 1189

Page 46: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

activity against a-glucosidase with IC50 values of 48–170 nM. It should be noted that

desulfated schulzeines A and B still retained activity (IC50 s 2.5 and 1.1 mM, respec-

tively) [152]. Recently, the stereochemistry at C-20 of 221was revised on the basis oftotal chemical synthesis [153].

O

N

O

O

OSO3Na

OSO3NaNaO3SO

penarolide sulfate A2 (233)

O

OSO3Na

N O

O

OSO3NaNaO3SO

penarolide sulfate A1 (232)

NH

HO OH

OH

235

N

OMe

O

O

OSO3Na

OSO3Na

penasulfate A (234)

HO

O

OH

callyspongynic acid (236)

23.4.10 S-Adenosylhomocysteine Hydrolase (EC 3.3.1.1)

S-adenosylhomocysteine hydrolase that reversibly hydrolyzes

S-adenosylhomocysteine to homocysteine and adenosine has been considered

an attractive target for the design of antiviral, antiparasitic, antiarthritic and immu-

nosuppressive agents. Ilimaquinone (237) has been reported to possess antiviral,

1190 Y. Nakao and N. Fusetani

Page 47: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

antimitotic, antimicrobial, and anti-HIV activities. It was also shown to vesiculate the

Golgi apparatus as well as to protect cells against the toxic effects of

ricin and diphtheria. Whole-cell and cytosolic photoaffinity labeling and affinity

chromatography studies with chemical probes prepared from 237 converged on

S-adenosylhomocysteine hydrolase as a cellular target of 237. Subsequent

studies with isolated enzyme indicated that ilimaquinone is a competitive

inhibitor of S-adenosylhomocysteine hydrolase (IC50 40 mM). On the basis of

SAR studies, pharmacophore modeling, and computer docking studies have allowed

for the design and preparation of a simplified structural variant (238) possessing a

100-fold greater inhibitory activity than that of natural (�)-ilimaquinone (237) [154].

O

O OMe

HO

H

O

HO OH

N

N

NN

NH2

O

HO OH

SO

O

NH3

N

N

NN

NH2

aliphaticregion

riboseregion

baseregion

S-adenosylhomocystein

(−)-ilimaquinone (237)

IC50 40 μM

238IC50 0.4 μM

23.4.11 Serine Proteases (EC 3.4.21.-)

Proteolytic enzymes are classified into serine proteases, cysteine proteases, aspartic

proteases, and metalloproteases on the basis of their catalytic centers. Trypsin-like

enzymes, a group of serine proteases, are associated with many disease states.

The hyperproteolytic activities of this homologous family of enzymes are attractive

chemotherapeutic targets in pathways of blood coagulation, fibrinolysis, kinin forma-

tion, complement activation, digestion, reproduction, and phagocytosis. Thus,

23 Enzyme Inhibitors from Marine Invertebrates 1191

Page 48: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

inhibitors of specific serine proteases can be potential drug-leads for many disease

status [155]. Cyclotheonamides A (239) and B (240) from a marine sponge Theonellasp. (now known to be T. swinhoei) inhibited thrombin, trypsin and plasmin with IC50

values of 0.076, 0.2, and 0.3 mg/mL, respectively [156]. Further investigation of

T. swinhoei yielded cyclotheonamides C (241) and D (242) [157]. Cyclotheonamides

E series (244–248) were not found from this sponge, but from the other type of

T. swinhoeiwhose interior part is white (E1–E3) [158] and from an Okinawan sponge

Ircinia sp. (E4–E5) [159]. Cyclotheonamides C (241), D (142), and E1–E5 (244–248)showed potent inhibition against thrombin and trypsin with IC50 values of 2.9–68 nM

and 7.4–55 nM, respectively. Cyclotheonamide E1 (244), E4 (247), and E5 (248)strongly inhibited both mouse and human tryptases (IC50s 6.9, 23, and 84.7 nM for

human tryptase, and 17.0, 6.5, and 54.1 nM for mouse tryptase, respectively) [159].

Dihydrocyclotheonamide A (243), pseudotheonamides A1 (249), A2 (250), B2 (251),C (252), andD (253) fromT. swinhoei, inwhich a-ketohomoarginine (k-Arg) residues

are cyclized, showed only moderate activity against these enzymes with IC50 values

ranging from 0.19 to 3.0 mM (thrombin) and 3.8 mM (trypsin) [160]. Complexes

between cyclotheonamide A and human a-thrombin and between cyclotheonamide A

and trypsin were successfully crystalized and their crystal structures were solved,

disclosing that cyclotheonamide A binds to the catalytic center of these enzyme;

particularly k-Argwhich forms the hemiacetal linkagewith Ser195 of one of a catalytic

triad is important for the binding to the enzyme [161]. From the same T. swinhoei, wasisolated a thrombin inhibitory linear peptide, nazumamide A (254) which inhibited

thrombin with an IC50 value of 2.8 mg/mL, but not trypsin at 100 mg/mL [162]. Retro-

binding mode of nazumamide A to human thrombin was demonstrated by X-ray

crystallography of the nazumamide A/thrombin complex [163].

HNH

O

OHN

N

HNO

HO

NH

O

HN

HN

H2N NH

O

O

OHN

NH

O

HN

HN

H2N

HN

NH

R

N

O

O

O

O

NH

OH

O

cyclotheonamide A (239) R = Hcyclotheonamide B (240) R = Ac

cyclotheonamide C (241)

O

1192 Y. Nakao and N. Fusetani

Page 49: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

O

O

O

R1 R2

O

O

E1 (244)

E2 (245)

E3 (246)

E4 (247)

E5 (248)

H

H

H

H

OH

NH

O

OHN

N

HNO

NH

O

HN

HN

H2N NH

O

O

OH

O

R1

HN

R2

cyclotheonamide E's

OHN

NH

HO

HN

HN

H2N

HN

NH

H

N

O

O

O

O

NH

OH

O

dihydrocyclotheonamide A (243)

HNH

O

OHN

N

HNO

NH

O

HN

HN

H2N NH

O

O

OH

cyclotheonamide D (242)

O

HN

ONH

O

HN

OHN

NHN

N

NH

O

OH

O

NHHO

H O

H

pseudotheonamide A2 (250)

HN

ONH

O

HN

OHN

NHN

N

NH

O

OH

O

NHHO

H O

H

pseudotheonamide A1 (249)

23 Enzyme Inhibitors from Marine Invertebrates 1193

Page 50: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

NH

O

HN

OHN

N NH2

O

OH

H O

O

NH2NH

N

HN

H2N NH

OOH

OH

O

HN

NH

OH

O

O

O

pseudotheonamide D (253) nazumamide A (254)

HHN

ON

O

N

OHN

NHN

N

NH

O

OH

O

NHHO

H O

pseudotheonamide B2 (251)

HN

N

NH

O

OH

O

NHHO

H O

O

NH2

NH

O

HN

OHN

N

pseudotheonamide C (252)

Toxadocials A–C (255–257) and toxadocic acid A (258) isolated from a marine

sponge Toxadocia sp., are alkyl sulfate thrombin inhibitors with IC50s of 6.5, 4.6,

3.2, and 2.7 mg/mL, respectively [164]. The mode of enzyme inhibition of

toxadocials is not known.

HOOSO3Na

OSO3Na OSO3Na OSO3Na OSO3Na

OSO3NaOSO3Na

OSO3NaOSO3Na

OSO3Na OSO3Na OSO3Na

HO

OH

OHHO

OHOOSO3Na OSO3Na OSO3NaOSO3Na

Toxadocial A (255)

Toxadocial B (256)

Toxadocial C (257)

Toxadocic acid A (258)

Dysinosins A–D (259–262) are inhibitors of factor VIIa and thrombin isolated

from the Australian sponge Lamellodysidea chlorea. Dysinosins showed potent

inhibitory activity against factor VIIa and thrombin with Ki values of 0.090 to 1.32

1194 Y. Nakao and N. Fusetani

Page 51: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

and 0.17 to -> 23 mM, respectively [165]. The structural similarity of dysinosins to

that of aeruginosins (263) which were the serine protease inhibitors isolated from

the cyanobactera Microcystis aeruginosa [166] and Oscillatoria agardhii [167]implied that they were biosynthesized by symbiotic microbes. Based on X-ray

crystallographic data of complexes of chlorodysinosin A with thrombin, a series

of analogues were synthesized varying the nature of the P1, P2, and P3pharmacophoric sites and the central octahydroindole carboxyamide core. Intro-

duction of a hydrophobic substituent on the D-Leu amide dramatically improved

the inhibitory activity against thrombin (264: IC50 1.6 nM) [168].

N

HN

HN

HO

H

HO

O

O

HO

OH

NHNH2

HN

OHaeruginosin 298-A (263)

N

HN

HN

H

HO

O

O

HO

NH2

HN

Cl

264

N

HN

HN

R2

R3

HO

R1

H

HO

O

O

MeO

N NH2

NH2

O

OHO

HOHOH2C

HO

R3R1

OH

OH

OH

dysinosine A (259)

B (260)

C (261)

D (262)

R2

OSO3−

OSO3−

OSO3−

OH

23 Enzyme Inhibitors from Marine Invertebrates 1195

Page 52: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

Factor XIa (FXIa) is a trypsin-like serine protease that plays a major role in the

amplification phase of the coagulation cascade and in maintaining clot integrity.

It is a unique target where specific inhibitors of FXIa might inhibit thrombosis

without completely interrupting the function of normal hemostasis and might

prevent or minimize the risk of hemostasis complication. Clavatadines A–E

(265–269) were isolated as FXIa inhibitors from the Australian sponge Subereaclavata along with known aerophobin 1 (270), purealdin L (271), and

aplysinamisine II (272). Compounds 265 and 266 inhibited FXIa with IC50 values

of 1.3 and 27 mM, respectively, whereas they did not inhibit FIXa (< 5% inhibition

at 222 mM). The crystal structure obtained by soaking of compound 265 into a singlecrystal of FXIa showed only the carbamate side chain of 265 covalently bonded to

the active site serine residue (Ser 195) [169]. Compounds 267–272 were shown to

be weaker inhibitors (12–59% inhibition at 222 mM) [170].

23.4.12 Cysteine Proteases (EC 3.4.22.-)

23.4.12.1 Cathepsin B (EC 3.4.22.1)Cysteine proteases which have cysteine and histidine as the catalytic center are

involved in cytosolic protein metabolisms. Cathepsin B, a cysteine protease, is

known to be involved in various disease states, such as inflammation, trauma,

muscular dystrophy, and tumors. In particular, its possible roles in cancer metastasis

are of major concern in cancer chemotherapy. Two peptidic inhibitors, tokaramide

A (273) [171] and miraziridine A (274) [172] with IC50 values of 29 and 1,400 ng/

mL were isolated from the marine sponge Theonella aff. mirabilis. Miraziridine

A (274) contains three unusual amino acid residues; particularly unusual are

vinylogous arginine and aziridine-2,3-dicarboxylic acid. Secobatzelline A (207)isolated from a sponge Batzella sp. showed inhibition against peptidase activity of

CPP32, a member of caspases which are cystein proteases playing amajor role in the

programmed cell death mechanism known as apoptosis, with an IC50 value of

0.02 mg/mL as well [130]. Asteropterin (275), a uniquemolecule with a combination

of lumazine and N-methyl histamine isolated from the marine sponge Asteropussimplex, inhibited cathepsin B with an IC50 value of 1.4 mg/mL [173].

OH

Br Br

OHN

R

O

O

NH

NH2

NH

265 : R = OH266 : R = NH2

O

Br Br

O

NHN

O

NH

NH2

NH

n

267 : n = 1268 : n = 2

1196 Y. Nakao and N. Fusetani

Page 53: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

O

Br Br

O

NHN

O

NH

NH2

NH

Me

HO

n

271 : n = 1272 : n = 2

OH

Br

NH

NHO

OHN NH2

NH

269

O

Br Br

O

NHN

O

Me

HO

270N

NH

HONH

NH

HN

NH

HN

OH

O O

O

OH O

O

NH

HN NH2

O

miraziridine A (274)

HN

NH

HN

H

NH

NH

HO

O

O

O

O

H2Ntokaramide A (273)

NH

HN

N

N

O

O

N

Me

NH

N

asteropterin (275)

23 Enzyme Inhibitors from Marine Invertebrates 1197

Page 54: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

23.4.13 Aspartic Proteases (EC 3.4.23.-)

23.4.13.1 HIV Protease (EC 3.4.23.47)Replication of HIV involves the expression of several viral polyproteins that

requires the presence of a virus-specific protease for their maturation. Inhibition

of this enzyme results in immature viral particles; therefore, HIV-1 protease is an

ideal target for AIDS chemotherapeutics.

Didemnaketals A (276) and B (277), isolated from a Palauan ascidianDidemnumsp., inhibited HIV-1 protease with IC50 values of 2 and 10 mM, respectively [174].

The unusual structures and potent activity of didemnaketals indicated their possi-

bility as attractive synthetic targets [175]. Recently, simplified analogues lacking

the spiroketal portion were synthesized, some of which were as potent as

didemnaketal A. It was also proposed that didemnaketals inhibit dimerization of

HIV-1 protease monomers [176].

MeO

O O OAc

O

O

O

O

OH

O

O

O

Odidemnaketal A (276)

MeO

O O OAc

O

O

O

O

OH

O

O

O

MeO

O

didemnaketal B (277)

23.4.14 Metalloproteases (EC 3.4.24.-)

Metalloproteases contain metal ions such as Zn2+, Co2+, Mn2+, etc., in their

catalytic centers. The matrix metalloproteinases (MMPs), members of a large

1198 Y. Nakao and N. Fusetani

Page 55: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

subfamily of proteases which contains a catalytic zinc-binding domain, play

important roles in the remodeling and degradation of extracellular matrix (ECM).

They are linked to a range of physiological and pathological processes, including

wound healing, bone remodeling, angiogenesis, inflammation, and tumor progres-

sion and metastasis. The membrane-type matrix metalloproteinases (MT-MMPs)

are known as key enzymes in tumor metastasis. They activate progelatinase A to the

fully matured form which in turn degrades type IV collagen, the major component

of the basal membrane which prevents tumor progression. Thus, inhibitors of MT-

MMPs are believed to be potential anticancer drugs. Ancorinosides B–D (279–281)[177] were isolated as MT1-MMP inhibitors from the marine sponge Penaressollasi along with known ancorinoside A (278) [178] which was originally isolated

as an inhibitor of blastulation of starfish embryos from a marine sponge Ancorinasp. Ancorinosides A–D (278–281) inhibited MT1-MMP with IC50 values of 440,

500, 370, and 180 mg/mL, respectively. Ancorinoside B (279) also inhibited

gelatinase A (MMP2) with an IC50 value of 22 mg/mL. Haplosamate A (65) andits congener (282) were isolated from a Japanese marine sponge Cribrochalina sp.

as MT1-MMP inhibitors with IC50 values of 150 and 160 mg/mL, respectively;

[179] 65 was originally isolated as an HIV integrase inhibitor from Philippine

haplosclerid sponges [32]. Ageladine A (283) is a fluorescent alkaloid isolated

from the marine sponge Agelas nakamurai. The IC50 values of 283 against

MMPs 1, 2, 8, 9, 12, and 13 were 1.2, 2.0, 0.39, 0.79, 0.33, and 0.47 mg/mL,

respectively, while its N-methylated derivatives did not inhibit MMP2. Unlike other

MMP inhibitors, ageladine A (283) was not capable to chelate Zn2+ and not

a competitive inhibitor against MMP2 [180]. With such interesting aspects of

inhibition mode, as well as its anti-angiogenic activity, ageladine A became

a target of total synthesis [181].

OO

OO

OH

N

OH

OH

COOH

HOOH

HOHO

OH

O

O

MeO

OO

OH

N

OH

OH

COOH

HO

O

O

MeO

OO

OHHO

HOOH

OO

OH

N

OH

OH

COOH

HO

O

O

MeO

OO

OHHO

HOOH

ancorinoside A (278)

ancorinoside B (279)

ancorinoside C (280)

23 Enzyme Inhibitors from Marine Invertebrates 1199

Page 56: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

O

H

H

H

OH

OHH

OP

O

OMeNaOOHNaO3SO

N

NNH

NH

Br

Br

H2N

282 ageladine A (283)

OO

OO

OH

N

OH

OH

COOH

HOOH

HOHO

OH

O

O

MeO

ancorinoside D (281)

23.4.15 Histone Deacetylases (EC 3.23.-)

Histone acetylation and deacetylation are catalyzed by histone acetyltransferases

(HATs) and histone deacetylases (HDACs), respectively, which play important

roles in transcriptional regulation [182]. Inhibitors of these enzymes are known to

induce cell cycle arrest [183], p53-independent induction of the cyclin dependent

kinase inhibitor p21 [184], tumor-selective apoptosis [185], and differentiation of

normal and malignant cells [186]. HDAC inhibitors were also demonstrated to

exert anti-angiogenic effects through the alteration of vascular endothelial

growth factor (VEGF) signaling [187]. These direct and indirect effects on

tumor growth and metastasis indicate that HDAC inhibitors are potential anti-

cancer agents. Six new psammaplins E–J (288–293) were isolated from the

marine sponge Pseudoceratina purpurea collected in Papua New Guinea along

with known psammaplins A–D (284–287), and bisaprasin (294). These com-

pounds showed potent inhibitory activity against HDAC (IC50s 2.1–327 nM)

and cell-based p21 promoter activity (IC50s 0.7–15 mM), in addition to inhibitory

activity against DNA methyltransferase (DNMT) [188]. NVP-LAQ824 (295)which was developed on the basis of structures of HDAC inhibitors including

psammaplins [189] entered phase I clinical trials in patients with solid tumors or

leukemia [190]. Three new cyclostellettamines, cyclostellettamine G (297),dehydrocyclostellettamines D (298) and E (299), were isolated together with

the known cyclostellettamine A (296) from a marine sponge of the genus

Xestospongia. These compounds inhibited HDAC with IC50 values of

17–80 mM [191]. Five new cyclic tetrapeptides, azumamides A–E (300–304)were isolated from the marine sponge Mycale izuensis. Azumamides showed

inhibitory activity against human HDAC at IC50 values of 0.045–1.3 mM. In the

cell-based assay using K562 cells, azumamide A (300) inhibited deacetylation

of Ac-H3 (Lys9 and Lys 14) and Ac-H4 (Lys 8) in a dose-dependent manner

1200 Y. Nakao and N. Fusetani

Page 57: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

(0.19–19 mM). Furthermore, azumamide A inhibited angiogenesis in the in vitro

vascular organization model using mouse ES cells [192].

OH

Br

O

NHO

NH

Y

S S

HN

NH2

O

O

E (288) Y =

SCN

SO2NH2

S S

HN OMe

O

A (284) Y =

B (285) Y =

C (286) Y =

D (287) Y =

S S

HN

N

O

OH

Br

OH

S S

HN

OH

O

O

S S

HN

OH

O

NNH2

S S

HN OEt

OSO2CH3

S S

HN

N

OO

OH

Br

OH

F (289) Y =

G (290) Y =

H (291) Y =

I (292) Y =

J (293) Y =

psammaplin

N

NH

OH

OH

NH

O

NVP-LAQ824 (295)

N

NH

S

HO

O

Br

OH

SNH

NOH

OH

O

Br

bisaprasin (294) 2

23 Enzyme Inhibitors from Marine Invertebrates 1201

Page 58: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

HNHN

HN

R4

R3

O

O

O

O

R1

O

HN

R2

R1

NH2

NH2

OH

NH2

OH

R2

H

OH

OH

H

H

R3 = R4

Me

Me

Me

HMe

azumamide A (300)

B (301)

C (302)

D (303)

E (304)

NN+

CF3COO−

CF3COO−

+

cyclostellettamine A (296) : m = 1, n = 2

G (297) : m = 1, n = 1

D (298) : m = 1, n = 4, Δ13

E (299) : m = 2, n = 4, Δ13dehydrocyclostellettamine

23.4.16 ATPases (EC 3.6.1.3)

23.4.16.1 H,K-ATPaseH,K-ATPase mediates acid secretion from gastric wall cells. Thus, excess activa-

tion of this enzyme leads to hyperacidity, causing gastric ulcer. In order to discover

potential antiulcer leads, a number of Japanese marine invertebrates have been

screened for inhibition of porcine H,K-ATPase which resulted in isolation of

several inhibitors.

S-(+)-curcuphenol (305) and dehydrocurcuphenol (306), isolated from

a Japanese marine sponge Epipolasis sp., inhibited porcine H,K-ATPase with

IC50s of 8.3 and 23 mM, respectively, while hydroxycurcuphenol was inactive

[193]. It should be noted that the R-(�)-curcuphenol had already been reported

from the Caribbean gorgonian Pseudopterogorgia rigida [194]. Hexaprenylhy-

droquinone sulfate (307) obtained from a marine sponge Dysidea sp. inhibited

1202 Y. Nakao and N. Fusetani

Page 59: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

H,K-ATPase with an IC50 of 4.6 mM [195]. Since 307 showed promising activity,

a number of its analogues were synthesized and evaluated for inhibition of acid

secretion and antiulcer activity, however, the results were disappointing [2].

Sinulamide (308) derived from a soft coral Sinularia sp. inhibited the enzyme

activity with an IC50 of 5.5 mM [2]. The structure of 308 was recently refined by

synthetic approach [196].

OH OH

S-(+)-curcuphenol (305) dehydrocurcuphenol (306)

H

OSO3Na

OH

6

hexaprenylhydroquinone sulafte (307)

H

NN NH3O

++

+

Cl−

Cl−

Cl−

3

sinulamide (308)

23.4.16.2 Na,K-ATPaseNa,K-ATPase regulates Na+ transport through cell membranes and is directly

related to contraction and relaxation of smooth muscles, thus indicating that

inhibitors of the enzyme are potential drug leads for cardiovascular diseases.

Agelasines A–F (309–314), 9-methyladenine derivatives of diterpenes, isolated

from the Okinawan marine sponge Agelas nakamurai, were reported to inhibit

Na,K-ATPase, though their detailed activity is not available [197]. Two hypotaur-

ocyamine derivatives of diterpene, agelasidines B (315) and C (316) from the same

sponge inhibited not only Na,K-ATPase with IC50 values of 10–50 mM, but also

contraction of smooth muscle [198].

23 Enzyme Inhibitors from Marine Invertebrates 1203

Page 60: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

H

N N

N

NH2N

+

Cl−

agelasin A (309)

H

N N

N

NH2N

+

Cl−

agelasin B (310)

H2N

H

N N

N

N

+

Cl−

agelasin C (311)

H2N

H

H

N N

N

N

+

Cl−

agelasin D (312)

N

N

N

N

+

H2N

Cl−

agelasin E (313)

N

N

N

N

H

+

H2N

Cl−agelasin F (314)

1204 Y. Nakao and N. Fusetani

Page 61: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

SNH

OO NH2

NH2

Cl−+

agelasidin B (315)

NH2

NH2

S

HN

O O+Cl−agelasidin C (316)

Iantheran A (317), its diacetate, and a-hydroxy enone derivatives, isolated from

an Australian marine sponge Ianthella sp. showed moderate inhibitory activity

against Na,K-ATPase with IC50 values of 2.5, 5.0, and 10 mM, respectively [199].

This sponge also yielded ianthesins A–D (318–321), of which ianthesins B-C

inhibited dog kidney Na,K-ATPase with IC50 values of 440, 50, and 280 mM,

respectively [200].

O

NHN O

OHO

Br

OMe

Br

HO

R

O

Br

Br

ianthesin A (318)

B (319)

D (321)

RβNMe2

αNMe2

NHSO3Na

O OSO3Na

NaO3SO

Br

Br

OH

O

Br

OH

Br

iantheran A (317)

HNO

Br

OMe

HOON

HN O

Br

HN

O

Br

Br

SO3Na

O NH

Br

Br

O OH

O

ON

OH

Br

OMe

Br

ianthesin C (320)

23 Enzyme Inhibitors from Marine Invertebrates 1205

Page 62: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

Xestoquinone (82), a polycyclic quinone, which was isolated together with the

known antimicrobial substance halenaquinone (79) [201], from the Okinawan

marine sponge Xestospongia sapra, inhibited Na,K-ATPase. Xestoquinone was

the first example of marine natural products having parallelism between the ino-

tropic action and Na,K-ATPase inhibition. Halenaquinol (80) [202], a natural

cardioactive pentacyclic hydroquinone, from the marine sponge Petrosia seriata,was found to be a powerful inhibitor of the rat brain stem and cortex Na,K-ATPases

and the rabbit muscle sarcoplasmic reticulum Ca2+-ATPase with IC50 values of

0.70, 1.3, and 2.5 mM, respectively [203]. Sarcochromenol sulfate A (322) andsarcohydroquinone sulfates A–C (325–327) isolated from the New Zealand sponge

Sarcotragus spinulosus inhibited the rat brain Na,K-ATPase with IC50 values of

1.6, 1.6, 1.4, and 1.3 mM, respectively, while sarcochromenol sulfates B (323) andC (324) were inactive [204].

OH

NaO3SO

H

OR

OSO3Na

sarcohydroquinone sulfate A (325)

B(326)

C (327)

RH

SO3Na

H

n

sarcochromenol sulfate A (322) n = 5B (323) n = 6C (324) n = 7

n

n6

7

8

23.4.16.3 Vacuolar ATPasesVacuolar H+-ATPases, a class of proton pumps present in all eukariotic cells, are

responsible for proton transport in bone-derived membrane vesicles, and the pro-

cess of osteoclast-mediated bone resorption is directly dependent on H+-

translocating ATPases. Inhibition of osteoclast vascular H+-ATPase may be effec-

tive for reducing the rate of bone resorption in pathological conditions such as

osteoporosis. Adociasulfate 1 (328), a sesterterpenoid isolated from an Australian

marine sponge Adocia sp., reduced proton pump activity in hen bone-derived

membrane vesicles with an IC50 value of 3.6 mM and proton pumping in brain-

derived vesicles with an IC50 value of 4.69 mM, while adociasulfates 7 (329) and8 (330) were less active (IC50 30 mM and 55% inhibition at 100 mM, respectively)

[205]. Recently, antitumor polyketides, salicylihalamides A (331) and B (332)isolated from a marine sponge Haliclona sp. [206] and lobotamides A–F (333–338) from the tunicate Aplidium lobatum [207], both of which share the core

structure with YM-75518 (339), an antifungal metabolite of Pseudomonas sp.

Q38009 [208], were found to be a potent and selective inhibitors of mammalian

V-ATPases (IC50s 0.40–14.0 nM) [209]. Palmerolide A (340) isolated from the

Antarctic tunicate Synoicum adareanum is a cytotoxic macrolide. It showed 3 orders

of magnitude greater sensitivity (LC50 18 nM) against melanoma relative to other

cell lines tested in the NCI 60 cell line panel.

1206 Y. Nakao and N. Fusetani

Page 63: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

Palmerolide A displayed a 60-cell-line activity profile which correlated, using

the NCI COMPARE algorithm, to several vacuolar ATPase inhibitors. Indeed, it

inhibited V-ATPase with an IC50 of 2 nM and to be active in NCI’s hollow-fiber

assay [210]. Following study on its stereochemistry by degradation [211] or total

synthesis [212] revised its stereochemistry at C7, C10, and C11. The SAR study of

palmerolides using synthetic analogues showed that a phenyl substitution on side

chain and removal of the C7 hydroxy group enhanced the potency, while the

selectivity against melanoma of 340 was not reproduced [213].

R1O

OR2

HOH

R1 R2

SO3Na SO3Na

OSO3Na

NaO3SO

H SO3Na

OH

H HH H

H H

adociasulfate 1 (328)

7 (329

adociasulfate 8 (330)

O

HN

OOH O

OH

salicylihalamide A (331) Δ17 trans

salicylihalamide B (332) Δ17 cis

O

OH

O

O

CH2R

O

HN

ON

OMe

OH

lobotamide A (333) R = H(= YM-75518A)lobotamide D (336) R = OH

O

OH

O

O

CH2R

O

HN

ON OH

MeO

lobotamide B (334) R = H

(= YM-75518B)

lobotamide E (337) R = OH

O

OH

O

O

CH2R

O

HN

OOH

NMeO

lobotamide C (335) R = H

(= YM-75518C)

lobotamide F (338) R = OH

23 Enzyme Inhibitors from Marine Invertebrates 1207

Page 64: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

O

OH

O

O

O

HN

OOH

N

OMe

YM-75518D (339)

O

OH

HO

O NH2

O

OHN

O

palmerolide A (340)

23.5 Lyases (EC 4.-.-.-)

23.5.1 ATP Citrate Lyase (EC 4.1.3.8)

Since the very-low-density lipoproteins (VLDL) are metabolic precursors of low-

density lipoproteins (LDL) which play a key role in hyperchoresterolemia,

intervention of VLDL synthesis has provided a strategic target for hypercholes-

terolemia therapy. Inhibitors of ATP-citrate lyase (ACL) are anticipated to

reduce the production of acetyl CoA and can affect both lipogenesis and

cholesterogenesis, and as a result, expected to reduce VLDL synthesis. Purpurone

(341), a poly catecholic pyrrole isolated from a marine sponge Iotrochota sp.,

inhibits ATP-citrate lyase in a dose-dependent manner with an IC50 value of

7 mM [214].

N

OHHO

HOOH

O O

OH

OH

HO

HO

OH

purpurone (341)

1208 Y. Nakao and N. Fusetani

Page 65: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

23.6 Isomerases (EC 5.-.-.-)

23.6.1 Topoisomerase

DNA topoisomerases are nuclear enzymes that catalyze DNA breaking and unwind-

ing during cellular replication and RNA transcription. In eukaryotic cells, DNA

topoisomerases I (topo I) and II (topo II) play distinct roles in DNA unwinding. Topo

I catalyzes single-strand “nicking” to allow supercoiled DNA to unwind, whereas

topo II facilitates chromosomal duplication by relaxing and unwinding the DNA

duplex. Thus, inhibitors of topo I and topo II are expected to control cancer cell

proliferation and thought to be important targets for cancer chemotherapy.

23.6.1.1 Topoisomerase I (EC 5.99.1.2)Xestoquinol sulfate (342) and xestosaprols A (343) and B (344), as well as a knowncompound 345 and halenaquinol sulfate (81), were isolated as topo I inhibitors fromthe Okinawan sponge Xestospongia sapra; 342–344 inhibited DNA topo I with

MICs of 10, 12.5, and 12.5 mg/mL, respectively, while 345, xestoquinone (82), andhalenaquinone (79) were more potent with MICs of 2.5, 2, and 0.4 mg/mL, respec-

tively [215]. Interestingly, halenaquinol sulfate (81) was not active in this assay.

Makaluvamine G (346), a cytotoxic pigment which was isolated from an Indone-

sian sponge Histodermella sp., inhibited topo I with an IC50 value of 3.0 mM [216].

Two ceramide 1-sulfates 347 and 348 from the Japanese bryozoa Watersiporacucullata inhibited human topo I with IC50s of 0.4 and 0.2 mM, respectively

[217]. Similarly, amphimic acids A (349) and related long-chain fatty acids from

an Australian sponge Amphimedon sp. showed topo I inhibition with IC50s ranging

from 0.47 to 6.7 mM [218].

O

OOH

OSO3Na

N

NHN

OH

OMe

Me+

makaluvamine G (346)

xestoquinol sulfate (342)

O

OR2

R1OH

H

xestosaprol A (343)xestosaprol B (344)

(345)

R1 = H, OH; R2 = OR1 = R2 = H, OHR1 = O; R2 = H, OH

23 Enzyme Inhibitors from Marine Invertebrates 1209

Page 66: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

−O3SO

−O3SO

HN

OH

O

HO

HN

OH

O

HO

347

348

HO

O

amphimic acid A (349)

23.6.1.2 Topoisomerase II (EC 5.99.1.3)Wakayin (350), a cytotoxic pyrroloiminoquinone alkaloid, isolated from the ascidian

Clavelina sp., inhibited topo II at 250 mM [219]. Pyrroloiminoquinones,

makaluvamines A–F (351–356) which were isolated from Fijian sponges of the

genus Zyzzya, showed topo II inhibition with IC50s of 41, >500, 420, 320, 310, and

25 mM, respectively, while related makaluvone and damirone B were not active. The

makaluvamines showed differential toxicity toward the topo II-sensitive CHO cell line

xrs-6; makaluvamines A (351) and C (353) exhibited in vivo antitumor activity against

the human ovarian carcinoma Ovcar3 implanted in athymic mice [220]. Similarly,

makaluvamineN (357) isolated from thePhilippine sponge Zyzzya fuliginosa exhibitedgreater than 90% inhibition of topo II at 5 mg/mL [221]. OthermakaluvaminesG–H did

not show inhibitory activity against topo II [222], whereas only makaluvamine

G showedmoderate inhibition against topo I [189].Bengacarboline (358), ab-carbolinealkaloid isolated fromamarine ascidianDidemnum sp., inhibited topoII at 32mM[223].

NR2

N

O

NH2

R1

+

R1 R2

NH

N

O

NH2

Me

+

makaluvamine B (352)

NH

HN

OHN

NH

+

makaluvamine A (351) Me

Me

H

makaluvamine C (353) Hwakayin (350)

1210 Y. Nakao and N. Fusetani

Page 67: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

NH

N

N

S

HN

O

N

N

HN

O

O

R

RH

MeS

RHdehydrokuanoniamine B (361)

kuanoniamine D (364)

cystodytin J (363)

diplamine (365)

NH

N

HN

O

R

HN

S

O

RHshermilamine B (359)

shermilamine C (362)

N

N

N

O

ascididemin (360)

NH

N

OHN

Me

S

Br

OH

+

makaluvamine F (356)

NH

HN

OHN

OH+

makaluvamine D (354)

makaluvamine E Δ (355)

NH

HN

O

NH2

Br

+

makaluvamine N (357)

NH

NH

NH

NH

H2N

bengacarboline (358)

23 Enzyme Inhibitors from Marine Invertebrates 1211

Page 68: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

Two aromatic alkaloids, shermilamine B (359) [224] and ascididemin (360) [225]were reported to inhibit topo II at 30 and 75 mM, respectively [226]. Similarly,

dehydrokuanoiamine B (361), shermilamine C (362), cystodytin (363), kuanoniamine

D (364), and diplamine (365) from an ascidian Diplosoma sp., inhibited topo II with

IC50 values of 115, 138, 8.4, 127, and 9.2 mM, respectively [227]

Popolohuanone E (366), an oxidatively dimerized arenarol derivative isolated

from a Pohnpei sponge Dysidea sp., was reported to be inhibitory against topo II

with an IC50 of 400 nM and selectively cytotoxic against the A549 non-small-cell

human lung cancer cell line (IC50 2.5 mg/mL) [228]. Puupehenone (367) and

21-chloro-puupehenone (368), sesquiterpene hydroquinones isolated from

a verongid sponge, were tested for a wide variety of biological activities including

inhibitory activity against topo II, adenosine deaminase (ADA), glutathione

reductase (GR), dihydrofolate reductase (DHFR), and thymidylate synthetase

(TS), which disclosed that 367 inhibited topo II with an IC50 value of 1 mg/mL,

in addition that 367 and 368 inhibited ADA at IC50s >25 and> 25 mg/mL,

respectively, GR with an IC50 of 6 mg/mL for 368, DHFR with IC50s of 5 and

5 mg/mL, respectively, and TS with IC50s of 8 and 3 mg/mL, respectively [229].

O

O

O

OH

HO

HO

H

H

O

OH

OX

H

H

popolohuanone E (366) puupehenone (367): X = H

21-chloro-puupehenone (368): X = Cl

HO

OH

Oelenic acid (369)

HN

O

NH

Br

Br

O

HO

Br O

NOH

Br OH

Br

NOH

O

bastadin 14 (371)

H

OH

OH

O

4

370

1212 Y. Nakao and N. Fusetani

Page 69: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

O

NNH

NH

O

N S

virenamide A (372)

Elenic acid (369), an unusual metabolite of an Indonesian sponge Plakinastrellasp., inhibited topo II with an IC50 value of 0.1 mg/mL [230], while 3-tetraprenyl-4-

hydroxy benzylic acid (370) isolated from themarine sponge Irciniamuscarum had an

IC50 of 0.5 mg/mL [231]. Bastadin 14 (371) from the marine sponge Psammaplysillapurpurea showed inhibitory activity against topo II and dehydrofolate reductase withIC50 values of 2.0 and 2.5 mg/mL, respectively [232]. VirenamideA (372), a cytotoxiclinear peptide from the Australian ascidian Diplosoma virens, exhibited topo II

inhibitory activity with an IC50 of 2.5 mg/mL [233].

23.7 Conclusions

More than 370 metabolites derived from marine invertebrates have been shown to

inhibit various enzymes. These enzymes play pivotal roles in many aspects of the

biological processes; therefore, inhibitors are useful for understanding complicated

biological phenomena. These inhibitors fall into a wide range of biogenetic classes,

ranging from simple terpenoids or polyketides to complex polyketides and

oligopeptides. Therefore, there are still good chance to find structurally unique

new enzyme inhibitors such as okadaic acid, calyculins, and cyclotheonamides.

However, at the same time, we should make efforts to find new activities of trivial

known compounds to maximize the potentials of marine natural products. As

mentioned in the introduction, one of the trends of marine natural products will

be the discovery of new biological activities.

While this chapter was being prepared, a huge earthquake (M 9.0) occurred at the

Northeastern part of Japan. Tsunami and nuclear pollution have damaged quite

seriously the coast and marine organisms of the wide areas. Since these areas are

bountiful of marine organisms, this damage will give considerable limitation to study

marine natural products from this part. We hope the environment will be recovered in

the near future.

23.8 Study Questions

1. Is there any room for new findings in marine natural products chemistry?

2. How far have we understood the potentials of marine natural products?

23 Enzyme Inhibitors from Marine Invertebrates 1213

Page 70: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

3. Which should we attach more importance, to new structures or to new activities?

4. What is the status of our understanding on the mechanisms of action for marine

natural products?

5. How can marine natural products chemistry contribute to the upcoming trend of

life sciences?

References

1. Umezawa H (1982) Low-molecular-weight enzyme-inhibitors of microbial origin. Annu

Rev Microbiol 36:75–99

2. Fusetani N (1990) Research toward drugs from the sea. New J Chem 14:721–728

3. (a) Sato A, Morishita T, Shiraki T, Yoshioka S, Horikoshi H, Kuwano H, Hanzawa H, Hata T

(1993) Aldose reductase inhibitors from a marine sponge Dictyodendrilla sp. J Org Chem

58:7632–7634; (b) Wang Z, Ling B, Zhang R, Suo Y, Liu Y, Yu Z, Liu C (2009) Docking

and molecular dynamics studies toward the binding of new natural phenolic marine inhib-

itors and aldose reductase. J Mol Graph Model 28:162–169

4. Tasdemir D, Topaloglu B, Perozzo R, Brun R, O’Neill R, Carballeira NM, Zhang X,

Tonge PJ, Linden A, R€uedi P (2007) Marine natural products from the Turkish sponge

Agelas oroides that inhibit the enoyl reductases from Plasmodium falciparum, Mycobacte-rium tuberculosis and Escherichia coli. Bioorg Med Chem 15:6834–6845

5. Shin J, Fenical W (1991) Fuscosides A-D: antiinflammatory diterpenoid glycosides of new

structural classes from the Caribbean gorgonian Eunicea fusca. J Org Chem 56:3153–3158

6. (a) Jacobson PB, Jacobs RS (1992) Fucoside – an antiinflammatorymarine natural product which

selectively inhibits 5-lipoxygenase. 1. Physiological and biochemical-studies in murine inflam-

matory models. J Pharmacol Exp Ther 262:866–873; (b) Jacobson PB, Jacobs RS (1992)

Fucoside – an antiinflammatorymarine natural productwhich selectively inhibits 5-lipoxygenase.

2. Biochemical-studies in the human neutrophil. J Pharmacol Exp Ther 262:874–882

7. Niwa H, Watanabe M, Inagaki H, Yamada K (1994) Didemnilactones A and B and

neodidemnilactone, 3 new fatty-acid metabolites isolated from the tunicate Didemnummoseleyi (Herdman). Tetrahedron 50:7385–7400

8. Brastianos HC, Vottero E, Patrick BO, van Soest R, Matainaho T, Mauk AG, Andersen RJ

(2006) Exiguamine A, an indoleamine-2,3-dioxygenase (IDO) inhibitor isolated from the

narine sponge Neopetrosia exigua. J Am Chem Soc 128:16046–16047

9. Carr G, Chung MKW, Mauk AG, Andersen RJ (2008) Synthesis of indoleamine 2,3-

dioxygenase inhibitory analogues of the sponge alkaloid exiguamine A. J Med Chem

51:2634–2637

10. Pereira A, Vottero E, Roberge M, Mauk AG, Andersen RJ (2006) Indoleamine 2,3-

dioxygenase inhibitors from the Northeastern Pacific marine hydroid Garveia annulata.J Nat Prod 69:1496–1499

11. Tapiolas DM, Bowden BF, Abou-Mansour E, Willis RH, Doyle JR, Muirhead AN, Liptrot C,

Llewellyn LE, Wolff CWW, Wright AD, Motti CA (2009) Eusynstyelamides A, B, and C,

nNOS inhibitors, from the ascidian Eusynstyela latericius. J Nat Prod 72:1115–1120

12. Shinoda K, Morishita Y, Sasaki K, Matsuda Y, Takahashi I, Nishi T (1997) Enzymatic

characterization of human alpha 1,3-fucosyltransferase Fuc-TVII synthesized in a B cell

lymphoma cell lines. J Biol Chem 272:31992–31997

13. Wakimoto T, Maruyama A, Matsunaga S, Fusetani N, Shinoda K, Murphy PT (1999) Octa-

and nonaprenylhydroquinone sulfates, inhibitors of alpha 1,3-fucosyltransferase VII, from an

Australian marine sponge Sarcotragus sp. Bioorg Med Chem Lett 9:727–730

14. Gray CA, de Lira SP, Silva M, Pimenta EF, Thiemann OH, Oliva G, Hajdu E, Andersen RJ,

Berlinck RGS (2006) Sulfated meroterpenoids from the Brazilian sponge Callyspongia sp.

1214 Y. Nakao and N. Fusetani

Page 71: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

are inhibitors of the antileishmaniasis target adenosine phosphoribosyl transferase. J Org

Chem 71:8685–8690

15. Coval SJ, Patton RW, Petrin JM, James L, Rothofsky ML, Lin SL, Patel M, Reed JK, McPhail

AT, Bishop WR (1996) A cembranolide diterpene farnesyl protein transferase inhibitor from

the marine soft coral Lobophytum cristagalli. Bioorg Med Chem Lett 6:909–912

16. Ledroit V, Debitus C, Ausseil F, Raux R, Menou JL, Hill BT (2004) Heteronemin as

a protein farnesyl transferase inhibitor. Pharm Biol 42:454–456

17. Mori D, Kimura Y, Kitamura S, Sakagami Y, Yoshioka Y, Shintani T, Okamoto T,

Ojika M (2007) Spongolactams, farnesyl transferase inhibitors from a marine sponge:

isolation through an LC/MS-guided assay, structures, and semisyntheses. J Org Chem 72:

7190–7198

18. Desoubzdanne D, Marcourt L, Raux R, Chevalley S, Dorin D, Doerig C, Valentin A, Ausseil

F, Debitus C (2008) Alisiaquinones and alisiaquinol, dual inhibitors of Plasmodiumfalciparum enzyme targets from a New Caledonian deep water sponge. J Nat Prod

71:1189–1192

19. Seo Y, Cho KW, Rho JR, Shin J, Kwon BM, Bok SH, Song JI (1996) Solandelactones A-I,

lactonized cyclopropyl oxylipins isolated from the hydroid Solanderia secunda. Tetrahedron52:10583–10596

20. Casey PJ, Thissen JA, Moomaw JF (1991) Enzymatic modification of proteins with

a geranylgeranyl isoprenoid. Proc Natl Acad Sci USA 88:8631–8635

21. Kondoh O, Tachibana Y, Ohya Y, Arisawa M, Watanabe T (1997) Cloning of the RHO1 gene

fromCandida albicans and its regulation of beta-1,3-glucan synthesis. J Bacteriol 179:7734–774122. Drgonova J, Drgon T, Tanaka K, Kollar R, Chen GC, Ford RA, Chan CSM, Takai Y, Cabib

E (1996) Rhol p, a yeast protein at the interface between cell polarization and morphogen-

esis. Science 272:277–279

23. (a) Li HY, Matsunaga S, Fusetani N (1994) Corticaticacids A-C, antifungal acetylenic acids

from the marine sponge, Petrosia corticata. J Nat Prod 57:1464–1467; (b) Nishimura S,

Matsunaga S, Shibazaki M, Suzuki K, Harada N, Naoki H, Fusetani N (2002) Corticatic

acids D and E, polyacetylenic geranylgeranyltransferase type I inhibitors, from the marine

sponge Petrosia corticata. J Nat Prod 65:1353–1356

24. Nishimura S, Matsunaga S, Shibazaki M, Suzuki K, van Furihata K, Soest RWM, Fusetani

N (2003) Massadine, a novel geranylgeranyltransferase type I inhibitor from the marine

sponge Stylissa aff. massa. Org Lett 5:2255–2257

25. Ishiyama H, Ishibashi M, Ogawa A, Yoshida S, Kobayashi J (1997) Taurospongin A, a novel

acetylenic fatty acid derivative inhibiting DNA polymerase beta and HIV reverse transcrip-

tase from sponge Hippospongia sp. J Org Chem 62:3831–3836

26. Jimenez C, Quinoa E, Adamczeski M, Hunter LM, Crews P (1991) Novel sponge-derived

amino acids. 12. Tryptophan-derived pigments and accompanying sesterterpenes from

Fascaplysinopis reticulata. J Org Chem 56:3403–3410

27. Isaacs S, Hizi A, Kashman Y (1993) Toxicols A-C and toxiusol – new bioactive hexaprenoid

hydroquinones from Toxiclona toxius. Tetrahedron 49:4275–4282

28. Rudi A, Yosief T, Loya S, Hizi A, Schleyer M, Kashman Y (2001) Clathsterol, a novel anti-

HIV-1 RT sulfated sterol from the sponge Clathria species. J Nat Prod 64:1451–1453

29. Loya S, Rudi A, KashmanY, Hizi A (1999) Polycitone A, a novel and potent general inhibitor

of retroviral reverse transcriptases and cellular DNA polymerases. Biochem J 344:85–92

30. (a) Reddy MVR, Rao MR, Rhodes D, Hansen MST, Rubins K, Bushman FD, Venkateswarlu

Y, Faulkner DJ (1999) Lamellarin a 20-sulfate, an inhibitor of HIV-1 integrase active against

HTV-1 virus in cell culture. J Med Chem 42:1901–1907; (b) Yamaguchi T, Fukuda T,

Ishibashi F, Iwao M (2006) The first total synthesis of lamellarin a 20-sulfate, a selective

inhibitor of HIV-1 integrase. Tetrahedron Lett 47:3755–3757

31. Mitchell SS, Rhodes D, Bushman FD, Faulkner DJ (2000) Cyclodidemniserinol trisulfate,

a sulfated serinolipid from the Palauan ascidian Didemnum guttatum that inhibits HIV-1

integrase. Org Lett 2:1605–1607

23 Enzyme Inhibitors from Marine Invertebrates 1215

Page 72: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

32. Qureshi A, Faulkner DJ (1999) Haplosamates A and B: new steroidal sulfamate esters from

two haplosclerid sponges. Tetrahedron 55:8323–8330

33. Bifulco G, Bruno I, Minale L, Riccio R, Debitus C, Bourdy G, Vassas A, Lavayre J (1995)

Bioactive prenylhydroquinone sulfates and a novel C31 furanoterpene alcohol sulfate from

the marine sponge, Ircinia sp. J Nat Prod 58:1444–1449

34. (a) Blackburn EH (2001) Switching and signaling at the telomere. Cell 106:661–673;

(b) Cech TR (2000) Life at the end of the chromosome: telomeres and telomerase. Angew

Chem Int Ed 39:34–43; (c) Maser RS, DePinho RA (2002) Connecting chromosomes, crisis,

and cancer. Science 297:565–569

35. Lavelle F, Riou JF, Laoui A, Mailliet P (2000) Telomerase: a therapeutic target for the third

millennium? Crit Rev Oncol Hematol 34:111–126

36. (a) White LK, Wright WE, Shay JW (2001) Telomerase inhibitors. Trends Biotechnol

19:114–120; (b) Neidle S, Parkinson G (2002) Telomere maintenance as a target for

anticancer drug discovery. Nat Rev Drug Discov 1:383–393; (c) Helder MN, Wisman

GBA, van der Zee AGJ (2002) Telomerase and telomeres: from basic biology to cancer

treatment. Cancer Invest 20:82–101

37. Gowan SM, Harrison JR, Patteson L, Valenti M, Read MA, Neidle S, Kelland LR

(2002) A G-quadruplex-interactive potent small-molecule inhibitor of telomerase exhibiting

in vitro and in vivo antitumor activity. Mol Pharmacol 61:1154–1162

38. Warabi K, Matsunaga S, van Soest RWM, Fusetani N (2003) Dictyodendrins A-E, the first

telomerase-inhibitory marine natural products from the sponge Dictyodendrillaverongiformis. J Org Chem 68:2765–2770

39. F€urstner A, Domostoj MM, Scheiper B (2006) Total syntheses of the telomerase inhibitors

dictyodendrin B, C, and E. J Am Chem Soc 128:8087–8094

40. Warabi K, Hamada T, Nakao Y, Matsunaga S, Hirota H, van Soest RWM, Fusetani N (2005)

Axinelloside A, an unprecedented highly sulfated lipopolysaccharide inhibiting telomerase,

from the marine sponge, Axinella infundibula. J Am Chem Soc 127:13262–13270

41. (a) Frampton JE, Easthope SE (2004) Gefitinib – A review of its use in the management of

advanced non-small-cell lung cancer. Drugs 64:2475–2492; (b) Ciardiello F (2000) Epider-

mal growth factor receptor tyrosine kinase inhibitors as anticancer agents. Drugs 60 (Suppl

1):25–32; (c) Baselga J, Averbuch SD (2000) ZD1839 (‘Iressa’)(1,2) as an anticancer agent.

Drugs 60 (Suppl 1):33–40

42. He H, Kulanthaivel P, Baker BJ, Kalter K, Darges J, Cofield D, Wolff L, Adams L (1995)

New antiproliferative and antiinflammatory 9,11-secosterols from the gorgonian

Pseudopterogorgia sp. Tetrahedron 51:51–58

43. Kobayashi J, Inaba K, Tsuda M (1997) Tauroacidins A and B, new bromopyrrole alkaloids

possessing a taurine residue from Hymeniacidon sponge. Tetrahedron 53:16679–16682

44. Kreuter MH, Leake RE, Rinaldi F, M€uller-Klieser W, Maidhof A, M€uller WEG, Schroder

HC (1990) Inhibition of intrinsic protein tyrosine kinase-activity of EGF-receptor kinase

complex from human breast cancer cells by the marine sponge metabolite (+)-aeroplysinin-

1. Comp Biochem Physiol 97B:151–158

45. Hirano K, Kubota T, Tsuda M, Watanabe K, Fromont J, Kobayashi J (2000) Ma’edamines

A and B, cytotoxic bromotyrosine alkaloids with a unique 2(1H)pyrazinone ring from

sponge Suberea sp. Tetrahedron 56:8107–8110

46. Lee RH, Slate DL, Moretti R, Alvi KA, Crews P (1992) Marine sponge polyketide inhibitors

of protein tyrosine kinase. Biochem Biophys Res Commun 184:765–772

47. Fujiwara H, Matsunaga K, Saito M, Hagiya S, Furukawa K, Nakamura H, Ohizumi Y (2001)

Halenaquinone, a novel phosphatidylinositol 3-kinase inhibitor from a marine sponge,

induces apoptosis in PC12 cells. Eur J Pharmacol 413:37–45

48. Alvi KA, Rodrıguez J, Diaz MC, Moretti R, Wilhelm RS, Lee RH, Slate DL, Crews

P (1993) Protein-tyrosine kinase inhibitory properties of planar polycyclics obtained

from the marine sponge Xestospongia cf carbonaria and from total synthesis. J Org

Chem 58:4871–4880

1216 Y. Nakao and N. Fusetani

Page 73: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

49. Alvi KA, Diaz MC, Crews P, Slate DL, Lee RH, Moretti R (1992) Evaluation of new

sesquiterpene quinones from two Dysidea sponge species as inhibitors of protein tyrosine

kinase. J Org Chem 57:6604–6607

50. Buchanan MS, Edser A, King G, Whitmore J, Quinn RJ (2001) Cheilanthane sesterterpenes,

protein kinase inhibitors, from a marine sponge of the genus Ircinia. J Nat Prod 64:300–303

51. Tasdemir D, Mallon R, Greenstein M, Feldberg LR, Kim SC, Collins K, Wojciechowics D,

Mangalindan GC, Concepcion GP, Harper MK, Ireland CM (2002) Aldisine alkaloids from

the Philippine sponge Stylissa massa are potent inhibitors of mitogen-activated protein

kinase kinase-1 (MEK-1). J Med Chem 45:529–532

52. (a) Curman D, Cinel B, Williams DE, Rundle N, Block WD, Goodarzi AA, Hutchins JR,

Clarke PR, Zhou BB, Lees-Miller SP, Andersen RJ, Roberge M (2001) Inhibition of the G(2)

DNA damage checkpoint and of protein kinases Chk1 and Chk2 by the marine sponge alkaloid

debromohymenialdisine. J Biol Chem 276:17914–17919; (b) Meijer L, Thunnissen AMWH,

White AW, Garnier M, Nikolic M, Tsai LH, Walter J, Cleverley KE, Salinas PC, Wu YZ,

Mandelkow EM, Kim SH, Pettit GR (2000) Inhibition of cyclin-dependent kinases, GSK-3

beta and CK1 by hymenialdisine, a marine sponge constituent. Chem Biol 7:51–63;

(c) Annoura H, Tatsuoka T (1995) Total synthesis of hymenialdisine and debromohyme-

nialdisine – stereospecific construction of the 2-amino-4-oxo-2-imidazolin-5(Z)-disubstitutedylidene ring-system. Tetrahedron Lett 36:413–416; (d) Xu YZ, Yakushijin K, Horne DA

(1997) Synthesis of C11N5 marine sponge alkaloids: (+/�)-hymenin, stevensine,

hymenialdisine, and debromohymenialdisine. J Org Chem 62:456–464; (e) Sosa ACB,

Yakushijin K, Horne DA (2000) A practical synthesis of (Z)-debromohymenialdisine. J Org

Chem 65:610–611; (f) Prager RH, Tsopelas C (1992) Knoevenagel reactions of 6,7-

dihydropyrrolo [2,3-c]azepine-4,8(1H, 5H)-dione: an approach to the synthesis of pyrrolic

marine natural products. Aust J Chem 45:1771–1777; (g) Cho H, Matsuki S, Mizuno A,

Annoura H, Tatsuoka T (1997) Synthesis of pyrroloazepines. Facile synthesis of 2-substituted

pyrrole derivatives by the phosgene method. J Heterocycl Chem 34:87–91; (h) Portevin B,

Golsteyn RM, Pierre A, De Nanteuil G (2003) An expeditious multigram preparation of the

marine protein kinase inhibitor debromohymenialdisine. Tetrahderon Lett 44:9263–9265

53. Wan YQ, Hur WY, Cho CY, Liu Y, Adrian FJ, Lozach O, Bach S, Mayer T, Fabbro D,

Meijer L, Gray NS (2004) Synthesis and target identification of hymenialdisine analogs.

Chem Biol 11:247–259

54. (a) Takahashi I, Saitoh Y, Yoshida M, Sano H, Nakano H, Morimoto M, Takamori T (1989)

UCN-01 and UCN-02, new selective inhibitors of protein kinase C. II. Purification, physi-

cochemical properties, structural determination and biological activities. J Antibiot 42:571–

576; (b) Koshino H, Osada H, Isono K (1992) A new inhibitor of protein kinase C, RK-1409

(7-oxostaurosporine). II. Fermentation, isolation, physicochemical properties and structure.

J Antibiot 45:195–198

55. Kinnel RB, Scheuer PJ (1992) 11-Hydroxystaurosporine: a highly cytotoxic, powerful

protein kinase C inhibitor from a tunicate. J Org Chem 57:6327–6329

56. Horton PA, Longley RE, McConnell OJ, Ballas LM (1994) Staurosporine aglycone (K252-

C) and srcyriaflavin-A from the marine ascidian, Eudistoma. Experientia 50:843–84557. (a) Rodrıguez J, Peters BM, Kurz L, Schatzman RC, McCarley D, Lou L, Crews P (1993) An

alkaloid protein kinase C inhibitor, xestocyclamine A, from the marine sponge Xestospongiasp. J Am Chem Soc 115:10436–10437; (b) Rodrıguez J, Crews P (1994) Novel marine

sponge alkaloids acids. 7. Revised structure of xestocyclamine A and description of a new

analogue. Tetrahedron Lett 35:4719–4722

58. Walz AJ, Sundberg RJ (2000) Synthesis of 8-methoxy-1-methyl-1H-benzo[de][1,6]naphthyridin-9-ol (isoaaptamine) and analogues. J Org Chem 65:8001–8010 and references

cited therein

59. Patil AD, Freyer AJ, Killmer L, Hofmann G, Johnson RK (1997) Z-axinohydantoin and

debromo-Z-axinohydantoin from the sponge Stylotella aurantium: inhibitors of protein

kinase C. Nat Prod Lett 9:201–207

23 Enzyme Inhibitors from Marine Invertebrates 1217

Page 74: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

60. Chan JA, Freyer AJ, Carte BK, Hemling ME, Hofmann GA, Mattern MR, Mentzer MA,

Westley JW (1994) Protein kinase C inhibitors: novel spirosesquiterpene aldehydes from

a marine sponge Aka (¼ Siphonodictyon) coralliphagum. J Nat Prod 57:1543–1548

61. (a) Shigemori H, Madono T, Sasaki T, Mikami Y, Kobayashi J (1994) Nakijiquinone A and

B, new antifungal sesquiterpenoid quinones with an amino acid residue from an Okinawan

marine sponge. Tetrahedron 50:8347–8354; (b) Kobayashi J, Madono T, Shigemori H (1995)

Nakijiquinone C and D, new sesquiterpenoid quinones with a hydroxy amino-acid residue

from a marine sponge inhibiting c-erbB-2 kinase. Teterahedron 51:10861–10874

62. (a) Stahl P, Kissau L, Mazitschek R, Huwe A, Furet P, Giannis A, Waldmann H (2001) Total

synthesis and biological evaluation of the nakijiquinones. J Am Chem Soc 123:11586–

11593; (b) Stahl P, Kissau L, Mazitschek R, Giannis A, Waldmann H (2002) Natural product

derived receptor tyrosine kinase inhibitors: identification of IGF1R, Tie-2, and VEGFR-3

inhibitors. Angew Chem Int Ed 41:1174–1178

63. Patil AD, Freyer AJ, Killmer L, Offen P, Carte B, Jurewicz AJ, Johnson RK

(1997) Frondosins, five new sesquiterpene hydroquinone derivatives with novel skeletons

from the sponge Dysidea frondosa: inhibitors of interleukin-8 receptors. Tetrahedron

53:5047–5060

64. He HY, Kulanthaivel P, Baker BJ (1994) New cytotoxic sesterterpenes from the marine

sponge Spongia sp. Tetrahedron Lett 35:7189–7192

65. Alvi KA, Jaspars M, Crews P, Strulovici B, Oto E (1994) Penazetidine A, an alkaloid

inhibitor of protein kinase C. Bioorg Med Chem Lett 4:2447–2450

66. Willis RH, DeVries DJ (1997) BRS1, a C30 bis-amino, bis-hydroxy polyunsaturated lipid

from an Australian calcareous sponge that inhibits protein kinase C. Toxicon 35:1125–1129

67. Kobayashi J, Doi Y, Ishibashi M (1994) Shimofuridin A, a nucleoside derivative

embranching an acylfucopyranoside unit isolated from the Okinawan marine tunicate

Aplidium multiplicatum. J Org Chem 59:255–257

68. (a) Pettit GR, Herald CL, Doubek DL, Herald DL (1982) Anti-neoplastic agents. 86.

Isolation and structure of bryostatin 1. J Am Chem Soc 104:6846–6848; (b) Pettit GR

(1996) Progress in the discovery of biosynthetic anticancer drugs. J Nat Prod 59:812–821

69. (a) Kortmansky J, Schwartz GK (2003) Bryostatin-1: a novel PKC inhibitor in clinical

development. Cancer Invest 21:924–936; (b) Etcheberrigaray R, Tan M, Dewachter I,

Kuiperi C, Van der Auwera I, Wera S, Qiao L, Bank B, Nelson TJ, Kozikowski AP,

Van Leuven F, Alkon DL (2004) Therapeutic effects of PKC activators in Alzheimer’s

disease transgenic mice. Proc Natl Acad Aci USA 101:11141–11146; (c) Wender PA,

Verma VA (2006) Design, synthesis, and biological evaluation of a potent, PKC selective,

B-ring analog of bryostatin. Org Lett 8:1893–1896

70. Kobayashi J, Suzuki M, Tsuda M (1997) Konbu’acidin A, a new bromopyrrole alkaloid with

cdk4 inhibitory activity from Hymeniacidon sponge. Tetrahedron 53:15681–15684

71. Inaba K, Sato H, Tsuda M, Kobayashi J (1998) Spongiacidins A-D, new bromopyrrole

alkaloids from Hymeniacidon sponge. J Nat Prod 61:693–695

72. Sato H, Tsuda M, Watanabe K, Kobayashi J (1998) Rhopaladins A � D, new indole

alkaloids from marine tunicate Rhopalaea sp. Tetrahedron 54:8687–8690

73. Killday KB, Yarwood D, Sills MA, Murphy PT, Hooper JNA, Wright AE (2001) Microxine,

a new cdc2 kinase inhibitor from the Australian marine spongeMicroxina species. J Nat Prod64:525–526

74. Meijer L, Thunnissen AMWH, White AW, Garnier M, Nikolic M, Tsai LH, Walter J,

Cleverley KE, Salinas PC, Wu YZ, Biernat J, Mandelkow EM, Kim SH, Pettit GR

(2000) Inhibition of cylin-dependent kinases, GSK-3b and CK1 by hymenialdisine,

a marine sponge constituent. Chem Biol 7:51–63

75. Meijer L, Skaltsounis AL, Magiatis P, Polychronopoulos P, Knockaert M, Leost M, Ryan

XP, Vonica CA, Brivanlou A, Dajani R, Crovace C, Tarricone C, Musacchio A, Roe SM,

Peart L, Greengard P (2003) GSK-3-selective inhibitors derived from Tyrian purple

indirubins. Chem Biol 10:1255–1266

1218 Y. Nakao and N. Fusetani

Page 75: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

76. Gompel M, Leost M, Joffe EBK, Puricelli L, Franco LH, Palermo J, Meijer L (2004)

Meridianins, a new family of protein kinase inhibitors isolated from the ascidian Aplidiummeridianum. Bioorg Med Chem Lett 14:1703–1707

77. Hamann M, Alonso D, Martın-Aparicio E, Fuertes A, Perez-Puerto MJ, Castro A, Morales S,

Navarro ML, del Monte-Millan M, Medina M, Pennaka H, Balaiah A, Peng J, Cook J,

Wahyuono S, Martınez A (2007) Glycogen synthase kinase-3 (GSK-3) inhibitory activity

and structure-activity relationship (SAR) studies of the manzamine alkaloids. Potential for

Alzheimer’s disease. J Nat Prod 70:1397–1405

78. de Silva ED, Scheuer PJ (1980) Manoalide, an antibiotic sesterterpenoid from the marine

sponge Luffariella variabilis (Polejaff). Tetrahedron Lett 21:1611–1614

79. (a) de Freitas JC, Blankemeier LA, Jacobs RS (1984) Invitro inactivation of the neurotoxic

action of b-bungarotoxin by the marine natural product, manoalide. Experientia 40:864–865;

(b) Lombardo D, Dennis EA (1985) Cobra venom phospholipase A2 inhibition by

manoalide – a novel type of phospholipase inhibitor. J Biol Chem 260:7234–7240

80. de Silva ED, Scheuer PJ (1981) Three new sesterterpenoid antibiotics from the marine

sponge Luffariella variabilis (Polejaff.). Tetrahedron Lett 22:3147–3150

81. Katsumura S, Han Q, Kadono H, Fujiwara S, Isoe S, Fujii S, Nishimura H, Ikeda K (1992)

Phospholipase A2 inhibition by manoalide: development of simple analogs and necessary

functional groups for inhibition. Bioorg Med Chem Lett 2:1263–1266

82. Albizati KF, Holman T, Faulkner DJ, Glaser KB, Jacobs RS (1987) Luffariellolide, an

antiinflammatory sesterterpene from the marine sponge Luffariella sp. Experientia 43:949–

950

83. Kernan MR, Faulkner DJ, Jacobs RS (1987) The luffariellins, novel antiinflammatory

sesterterpenes of chemotaxonomic importance from the marine sponge Luffariella-variabilis. J Org Chem 52:3081–3083

84. (a) Potts BCM, Faulkner DJ, de Carvalho MS, Jacobs RS (1992) Chemical mechanism of

inactivation of bee venom phospholipase A2 by the marine natural-products manoalide,

luffariellolide, and scalaradial. J Am Chem Soc 114:5093–5100; (b) Fujii S, Tahara Y,

Toyomoto M, Hada S, Nishimura H, Inoue S, Ikeda K, Inagaki Y, Katsumura S, Samejima Y,

Omori-Satoh T, Takasaki C, Hayashi K (1995) Chemical modification and inactivation of

phospholipases A2 by a manoalide analog. Biochem J 308:297–304

85. Tanaka K, Kamatani M, Mori H, Fujii S, Ikeda K, Hisada M, Itagaki Y, Katsumura S (1999)

The inhibitory mechanism of bovine pancreatic phospholipase A2 by aldehyde terpenoids.

Tetrahedron 55:1657–1686

86. (a) De Rosa S, De Stefano S (1988) Cacosponginolide: a new antitumoral sesterterpene, from

the marine sponge Cacospongia mollior. J Org Chem 53:5020–5023; (b) Puliti R, De Rosa S,

Mattia CA, Mazzarella L (1990) Structure and stereochemistry of an acetate derivative of

cacospongionolide, a new antitumoral sesterterpenoid from marine sponge Cacospongiamollior. Acta Cryst C46:1533–1536; (c) De Rosa S, Crispino A, De Giulio A, Iodice C,

Pronzato R, Zavodnik N (1995) Cacospongionolide B, a new sesterterpene from the sponge

Fasciospongia cavernosa. J Nat Prod 58:1776–1780; (d) Soriente A, Crispino A, De Rosa M,

De Rosa S, Scettri A, Scognamiglio G, Villano R, Sodano G (2000) Stereochemistry of

antiinflammatory marine sesterterpenes. Eur J Org Chem:947–953

87. Pastor PG, De Rosa S, De Giulio A, Paya M, Alcaraz MJ (1999) Modulation of acute and

chronic inflammatory processes by cacospongionolide B, a novel inhibitor of human syno-

vial phospholipase A2. Brit J Pharmacol 126:301–311

88. De Rosa S, Crispino A, De Giulio A, Iodice C, Benrezzouk R, Terencio MC, Ferrandiz ML,

Alcaraz MJ, Paya M (1998) A new cacospongionolide inhibitor of human secretory phos-

pholipase A2 from the Tyrrhenian sponge Fasciospongia cavernosa and absolute configura-

tion of cacospongionolides. J Nat Prod 61:931–935

89. Cheung AK, Snapper ML (2002) Total syntheses of (+)- and (�)-cacospongionolide B: new

insight into structural requirements for phospholipase A2 inhibition. J Am Chem Soc

124:11584–11585

23 Enzyme Inhibitors from Marine Invertebrates 1219

Page 76: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

90. Posadas I, De Rosa S, Terencio MC, Paya M, Alcaraz MJ (2003) Cacospongionolide

B suppresses the expression of inflammatory enzymes and tumour necrosis factor-a by

inhibiting nuclear factor-k B activation. Brit J Pharm 138:1571–1579

91. Posadas I, Terencio MC, De Rosa S, Paya M (2000) Cavernolide: a new inhibitor of human

sPLA2 sharing unusual chemical features. Life Sci 67:3007–3014

92. Giannini C, Debitus C, Posadas I, Paya M, D’auria MV (2000) Dysidotronic acid, a new and

selective human phospholipase A2 inhibitor from the sponge Dysidea sp. Tetrahedron Lett

41:3257–3260

93. Giannini C, Debitus C, Lucas R, Ubeda A, Paya M, Hooper JNA, D’Auria MV

(2001) New sesquiterpene derivatives from the sponge Dysidea species with a selective

inhibitor profile against human phospholipase A2 and other leukocyte functions. J Nat Prod

64:612–615

94. Randazzo A, Debitus C, Minale L, Pastor PG, Alcaraz MJ, Paya M, Gomez-Paloma L (1998)

Petrosaspongiolides M-R: new potent and selective phospholipase A2 inhibitors from the

New Caledonian marine sponge Petrosaspongia nigra. J Nat Prod 61:571–575

95. (a) Dal Piaz F, Casapullo A, Randazzo A, Riccio R, Pucci P, Marino G, Gomez-Paloma L

(2002) Molecular basis of phospholipase A2 inhibition by petrosaspongiolide M.

ChemBioChem 3:664–671; (b) Monti MC, Casapullo A, Riccio R, Gomez-Paloma L

(2004) Further insights on the structural aspects of PLA2 inhibition by g-hydroxybu-tenolide-containing natural products: a comparative study on petrosaspongiolides M-R.

Bioorg Med Chem 12:1467–1474

96. Monti MC, Casapullo A, Riccio R, Gomez-Paloma L (2004) PLA2-mediated catalytic

activation of its inhibitor 25-acetyl-petrosaspongiolide M: serendipitous identification of

a new PLA2 suicide inhibitor. FEBS Lett 578:269–274

97. Monti MC, Riccio R, Casapullo A (2009) Effects of petrosaspongiolide R on the surface

topology of bee venom PLA2: a limited proteolysis and mass spectrometry analysis. Bioorg

Chem 37:6–10

98. Posadas I, Terencio MC, Randazzo A, Gomez-Paloma L, Paya M, Alcaraz MJ (2003) Inhi-

bition of the NF-k B signaling pathway mediates the anti-inflammatory effects of petrosas-

pongiolide M. Biochem Pharmacol 65:887–895

99. Crews P, Jimenez C, O’Neil-Johnson M (1991) Using spectroscopic and database strategies

to unravel structures of polycyclic bioactive marine sponge sesterterpenes. Tetrahedron

47:3585–3600

100. Kernan MR, Faulkner DJ, Parkanyi L, Clardy J, de Carvalho MS, Jacobs RS

(1989) Luffolide, a novel anti-inflammatory terpene from the sponge Luffariella sp.

Experientia 45:388–390

101. Alvi KA, Crews P (1992) Homoscalarane sesterterpnes from Lendenfeldia frondosa. J NatProd 55:859–865

102. Fontana A, Mollo E, Ortea J, Gavagnin M, Cimino G (2000) Scalarane and homoscalarane

compounds from the nudibranchs Glossodoris sedna and Glossodoris dalli: chemical and

biological properties. J Nat Prod 63:527–530

103. Cimino G, DeStefano S, Minale L (1974) Scalaradial, a sesterterpene with the tetracar-

bocyclic skeleton of scalarin, from the sponge Cacospongia mollior. Experientia 30:846–847104. Kernan MR, Faulkner DJ (1988) Sesterterpene sulfates from a sponge of the family

Halichondriidae. J Org Chem 53:4574–4578

105. Alfano G, Cimino G, DeStefano S (1979) Palinurin, a new linear sesterterpene from a marine

sponge. Experientia 35:1136–1137

106. Schmidt EW, Faulkner DJ (1996) Palauolol, a new anti-inflammatory sesterterpene from the

sponge Fascaplysinopsis sp. from Palau. Tetrahedron Lett 37:3951–3954

107. Shin JH, Seo YW, Rho JR, Cho KW (1996) Isolation of polyhydroxysteroids from the

gorgonian Acabaria undulata. J Nat Prod 59:679–682

108. Seo Y, Cho KW, Chung H, Lee HS, Shin J (1998) New secosteroids from a gorgonian of the

genus Muricella. J Nat Prod 61:1441–1443

1220 Y. Nakao and N. Fusetani

Page 77: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

109. Sepcic K, Guella G, Mancini I, Pietra F, Serra MD, Menestrina G, Tubbs K, Macek P, Turk

T (1997) Characterization of anticholinesterase-active 3-alkylpyridinium polymers from the

marine sponge Reniera sarai in aqueous solutions. J Nat Prod 60:991–996

110. Tachibana K, Scheuer PJ, Tsukitani Y, Kikuchi H, Engen VD, Clardy J, Gopichand Y,

Schmitz FJ (1981) Okadaic acid, a cytotoxic polyether from two marine sponges of the genus

Halichondria. J Am Chem Soc 103:2469–2471

111. Takai A, Murata M, Torigoe K, Isobe M, Mieskes G, Yasumoto Y (1992) Inhibitory effect of

okadaic acid derivatives on protein phosphatases: A study on structure-affinity relationship.

Biochem J 284:539–544

112. Maynes JT, Bateman KS, Chemey MM, Das AK, Luu HA, Holmes CF, James MN

(2001) Crystal structure of the tumor-promoter okadaic acid bound to protein phosphatase-

1. J Biol Chem 276:44078–44082

113. Cruz PG, Daranas AH, Fernandez JJ, Norte M (2007) 19-epi -Okadaic acid, a novel proteinphosphatase inhibitor with enhanced selectivity. Org Lett 9:3045–3048

114. Cruz PG, Fernandez JJ, Norte M, Daranas AH (2008) Belizeanic acid: a potent protein

phosphatase 1 inhibitor belonging to the okadaic acid class, with an unusual skeleton. Chem

Eur J 14:6948–6956

115. Kato Y, Fusetani N, Matsunaga S, Hashimoto K (1986) The bioactive marine metabolites.

16. Calyculin A, a novel antitumor metabolite from the marine sponge Discodermia calyx.J Am Chem Soc 108:2780–2781

116. Ishihara H, Martin BL, Brautigan DL, Karaki H, Ozaki H, Kato Y, Fusetani N, Watabe S,

Hashimoto K, Uemura D, Hartshorne DJ (1989) Calyculin A and okadaic acid: inhibitors of

protein phosphatase activity. Biochem Biophys Res Commun 159:871–877

117. (a) Kato Y, Fusetani N, Matsunaga S, Hashimoto K (1988) Isolation and structure elucida-

tion of calyculin B, C, and D, novel antitumor metabolites, from the marine sponge

Discodermia calyx. J Org Chem 53:3930–3932; (b) Matsunaga S, Fujiki H, Sakata D,

Fusetani N (1991) Calyculins E, F, G, and H, additional inhibitors of protein phosphatase

1 and 2A, from the marine sponge Discodermia calyx. Tetrahedron 47:2999–3006

118. (a) Matsunaga S, Wakimoto T, Fusetani N (1997) Bioactive marine metabolites. 78. Isolation

of four new calyculins from the marine sponge Discodermia calyx. J Org Chem

62:2640–2642; (b) Matsunaga S, Wakimoto T, Fusetani N, Suganuma M (1997) Isolation

of dephosphonocalyculin A from the marine sponge, Discodermia calyx. Tetrahedron Lett

38:3763–3764

119. Wakimoto T, Matsunaga S, Takai A, Fusetani N (2002) Insight into binding of calyculin A to

protein phosphatase 1: isolation of hemicalyculin A and chemical transformation of

calyculin A. Chem Biol 9:309–319

120. Kita A, Matsunaga S, Takai A, Kataiwa H, Wakimoto T, Fusetani N, Isobe M, Miki K (2002)

Crystal structure of the complex between calyculin A and the catalytic subunit of protein

phosphatase 1. Structure 10:715–724 & 1149

121. (a) Goldberg J, Huang HB, Kwon YG, Greengard P, Naim AC, Kuriyan J (1995) Three-

dimensional structure of the catalytic subunit of protein serine/threonine phosphatase-1.

Nature 376:745–753; (b) Egloff MP, Cohen PTW, Reinemer P, Barford D (1995) Crystal-

structure of the catalytic subunit of human protein phosphatase-1 and its complex with

tungstate. J Mol Biol 254:942–959

122. Fu X, Schmitz FJ, Kelly-Borges M, Mccready TL, Holmes CFB (1998) Clavosines A-C from

the marine sponge Myriastra clavosa: potent cytotoxins and inhibitors of protein phospha-

tases 1 and 2A. J Org Chem 63:7957–7963

123. de Silva ED, Williams DE, Andersen RJ, Klix H, Holmes CFB, Allen TM (1992) Motuporin,

a potent protein phosphatase inhibitor isolated from the Papua New Guinea sponge

Theonella swinhoei Gray. Tetrahedron Lett 33:1561–1564

124. Capon RJ, Rooney F, Murray LM, Collins E, Sim ATR, Rostas JAP, Butler MS, Carroll AR

(1998) Dragmacidins: new protein phosphatase inhibitors from a southern Australian deep-

water marine sponge, Spongosorites sp. J Nat Prod 61:660–662

23 Enzyme Inhibitors from Marine Invertebrates 1221

Page 78: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

125. Williams DE, Roberge M, Van Soest R, Andersen RJ (2003) Spirastrellolide A, an antimi-

totic macrolide isolated from the Caribbean marine sponge Spirastrella coccinea. J Am

Chem Soc 125:5296–5297

126. Williams DE, Lapawa M, Feng XD, Tarling T, Roberge M, Andersen RJ

(2004) Spirastrellolide A: revised structure, progress toward the relative configuration, and

inhibition of protein phosphatase 2A. Org Lett 6:2607–2610

127. Endo T, Tsuda M, Okada T, Mitsuhashi S, Shima H, Kikuchi K, Mikami Y, Fromont J,

Kobayashi J (2004) Nagelamides A-H, new dimeric bromopyrrole alkaloids from marine

sponge Agelas species. J Nat Prod 67:1262–1267

128. Guerini D (1997) Calcineurin: not just a simple protein phosphatase. Biochem Biophys Res

Comm 235:271–275

129. Liu J, Farmer JD, Lane WS, Friedman J, Weissman I, Schreiber SL (1991) Calcineurin is

a common target of cyclophilin-cyclosporine A and FKBP-FK506 complex. Cell 66:807–815

130. Gunasekera SP, McCarthy PJ, Longley RE, Pomponi SA, Wright AE (1999) Secobatzellines

A and B, two new enzyme inhibitors from a deep-water Caribbean sponge of the genus

Batzella. J Nat Prod 62:1208–1211

131. Gunasekera SP, McCarthy PJ, Kelly-Borges M, Lobkovsky E, Clardy J (1996) Dysidiolide:

a novel protein phosphatase inhibitor from the Caribbean sponge Dysidea etheria de

Laubenfels. J Am Chem Soc 118:8759–8760

132. (a) Corey EJ, Roberts BE (1997) Total synthesis of dysidiolide. J Am Chem Soc 119:12425–

12431; (b) Boukouvalas J, Cheng YX, Robichaud J (1998) Total synthesis of (+)-dysidiolide.

J Org Chem 63:228–229; (c) Magnuson SR, Sepp-Lorenzino L, Rosen N, Danishefsky SJ

(1998) A concise total synthesis of dysidiolide through application of a dioxolenium-

mediated Diels-Alder reaction. J Am Chem Soc 120:1615–1616; (d) Takahashi M, Dodo

K, Sugimoto Y, Aoyagi Y, Yamada Y, Hashhimoto Y, Shirai R (2000) Synthesis of the novel

analogues of dysidiolide and their structure-activity relationship. Bioorg Med Chem Lett

10:2571–2574; (e) Demeke D, Forsyth CJ (2000) Novel total synthesis of the anticancer

natural product dysidiolide. Org Lett 2:3177–3179; (f) Brohm D, Metzger S, Bhargava A,

M€uller O, Lieb F, Waldmann H (2002) Natural products are biologically validated starting

points in structural space for compound library development: solid-phase synthesis of

dysidiolide-derived phosphatase inhibitors. Angew Chem Int Ed 41:307–311

133. (a) Loukaci A, Le Saout I, Samadi M, Leclerc S, Damiens E, Meijer L, Debitus C, Guyot

M (2001) Coscinosulfate, a CDC25 phosphatase inhibitor from the sponge Coscinodermamathewsi. Bioorg Med Chem 9:3049–3054; (b) Poigny S, Nouri S, Chiaroni A, Guyot M,

Samadi M (2001) Total synthesis and determination of the absolute configuration of

coscinosulfate. A new selective inhibitor of Cdc25 protein phosphatase. J Org Chem

66:7263–7269

134. Berridge MJ (1987) Inositol trisphosphate as a 2nd messenger in signal transduction. Annu

Rev Biochem 56:159–193

135. Fukami A, Ikeda Y, Kondo S, Naganawa H, Takeuchi T, Furuya S, Hirabayashi Y, Shimoike

K, Hosaka S, Watanabe Y, Umezawa K (1997) Akaterpin, a novel bioactive triterpene from

the marine sponge Callyspongia sp. Tetrahedron Lett 38:1201–1202

136. Kobayashi J, Nakamura H, Ohizumi Y, Hirata Y (1986) Eudistomidin A, a novel

calmodulin antagonist from the Okinawan tunicate Eudistoma glaucus. Tetrahedron Lett

27:1191–1194

137. Kobayashi J, Cheng JF, Ohta T, Nozoe S, Ohizumi Y, Sasaki T (1990) Eudistomidins B, C,

and D: novel antileukemic alkaloids from the Okinawan marine tunicate Eudistoma glaucus.J Org Chem 55:3666–3670

138. (a) Ishibashi M, Ohizumi Y, Sasaki T, Nakamura H, Hirata Y, Kobayashi J (1987)

Pseudodistomins A and B, novel antineoplastic piperidine alkaloids with calmodulin antag-

onistic activity from the Okinawan tunicate Pseudodistoma kanoko. J Org Chem 52:450–

453; (b) Knapp S, Hale JJ (1993) Synthesis of (+)-tetrahydropseudodistomin. J Org Chem

58:2650–2651

1222 Y. Nakao and N. Fusetani

Page 79: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

139. Kobayashi J, Cheng JF, Kikuchi Y, Ishibashi M, Yamamura S, Ohizumi Y, Ohta T, Nozoe

S (1990) Rigidin, a novel alkaloid with calmodulin antagonistic activity from the Okinawan

marine tunicate Eudistoma cf rigida. Tetrahedron Lett 31:4617–4620

140. Hirota H, Matsunaga S, Fusetani N (1990) Bioactive marine metabolites. 32. Stellettamide A,

an antifungal alkaloid from a marine sponge of the genus Stelletta. Tetrahedron Lett

31:4163–4164

141. Abe Y, Saito S, Hori M, Ozaki H, Fusetani N, Karaki H (1997) Stellettamide-A, a novel

inhibitor of calmodulin, isolated from a marine sponge. Brit J Pharmacol 121:1309–1314

142. Tsukamoto S, Yamashita T, Matsunaga S, Fusetani N (1999) Bioactive marine metabolites –

Part 89 – Stellettazole A: an antibacterial guanidinoimidazole alkaloid from a marine sponge

Stelletta sp. Tetrahedron Lett 40:737–738

143. Tsukamoto S, Yamashita T, Matsunaga S, Fusetani N (1999) Bistellettadines A and B: two

bioactive dimeric stellettadines from a marine sponge Stelletta sp. J Org Chem 64:3794–3795

144. Nakao Y, Takada K, Matsunaga S, Fusetani N (2001) Calyceramides A-C: neuraminidase

inhibitory sulfated ceramides from the marine sponge Discodermia calyx. Tetrahedron57:3013–3017

145. Takada K, Nakao Y, Matsunaga S, van Soest RWM, Fusetani N (2002) Nobiloside, a new

neuraminidase inhibitory triterpenoidal saponin from the marine sponge Erylus nobilis. J NatProd 65:411–413

146. Takada K, Hamada T, Hirota H, Nakao Y, Matsunaga S, van Soest RWM, Fusetani N (2006)

Asteropine A, a sialidase-inhibiting conotoxin-like peptide from the marine sponge

Asteropus simplex. Chem Biol 13:569–574

147. Kato T, Shizuri Y, Izumida H, Yokoyama A, Endo M (1995) Styloguanidines, new

chitinase inhibitors from the marine sponge Stylotella aurantium. Tetrahedron Lett

36:2133–2136

148. Nakao Y, Maki T, Matsunaga S, van Soest RWM, Fusetani N (2000) Penarolide sulfates A1

and A2, new a-glucosidase inhibitors from a marine sponge Penares sp. Tetrahedron

56:8977–8987

149. Nakao Y, Maki T, Matsunaga S, van Soest RWM, Fusetani N (2004) Penasulfate A, a new

a-glucosidase inhibitor from a marine sponge Penares sp. J Nat Prod 67:1346–1350

150. Saludes JP, Lievens SC, Molinski TF (2007) Occurrence of the a-glucosidase inhibitor 1,4-dideoxy-1,4-imino-D-arabinitol and related iminopentitols in marine sponges. J Nat Prod

70:436–438

151. Nakao Y, Uehara T, Matsunaga S, Fusetani N, van Soest RWM (2007) Callyspongynic acid,

a polyacetylenic acid which inhibits a-glucosidase, from the marine sponge Callyspongiatruncata. J Nat Prod 65:922–924

152. Takada K, Uehara T, Nakao Y, Matsunaga S, van Soest RWM, Fusetani N (2004)

Schulzeines A-C, new a-glucosidase inhibitors from the marine sponge Penares schulzei.J Am Chem Soc 126:187–193

153. Bowen EG, Wardrop DJ (2009) Total synthesis of the a-glucosidase inhibitors schulzeine A,B, and C and a structural revision of schulzeine A. J Am Chem Soc 131:6062–6063

154. Kim BG, Chun TG, Lee HY, Snapper ML (2009) A new structural class of

S-adenosylhomocysteine hydrolase inhibitors. Bioorg Med Chem 17:6707–6714

155. Bertrand JA, Oleksyszyn J, Kam CM, Boduszek B, Presnell S, Plaskon RR, Suddath FL,

Powers JC, Williams LD (1996) Inhibition of trypsin and thrombin by amino(4-

amidinophenyl)methanephosphonate diphenyl ester derivatives: X-ray structures and molec-

ular models. Biochemistry 35:3147–3155

156. Fusetani N, Matsunaga S, Matsumoto H, Takebayashi Y (1990) Bioactive marine metabo-

lites. 33. Cyclotheonamides, potent thrombin inhibitors, from a marine sponge Theonella sp.J Am Chem Soc 112:7053–7054

157. Nakao Y, Matsunaga S, Fusetani N (1995) Three more cyclotheonamides C, D, and E,

potent thrombin inhibitors from the marine sponge Theonella swinhoei. Bioorg Med Chem

3:1115–1122

23 Enzyme Inhibitors from Marine Invertebrates 1223

Page 80: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

158. Nakao Y, Oku N, Matsunaga S, Fusetani N (1998) Cyclotheonamides E2 and E3, new potent

serine protease inhibitors from the marine sponge of the genus Theonella. J Nat Prod 61:

667–670

159. Murakami Y, Takei M, Shindo K, Kitazume C, Tanaka J, Higa T, Fukamachi H (2002)

Cyclotheonamide E4 and E5, new potent tryptase inhibitors from an Ircinia species of

sponge. J Nat Prod 65:259–261

160. Nakao Y, Masuda A, Matsunaga S, Fusetani N (1999) Pseudotheonamides, serine protease

inhibitors from the marine sponge Theonella swinhoei. J Am Chem Soc 121:2425–2431

161. (a) Lee AY, Hagihara M, Karmacharya R, Albers MW, Schreiber SL, Clardy J (1993)

Atomic structure of the trypsin cyclotheonamide A complex: lessons for the design of

serine protease inhibitors. J Am Chem Soc 115:12619–12620; (b) Maryanoff BE, Qiu

XY, Padmanabhan KP, Tulinsky A, Almond Jr HR, Andrade-Gordon P, Greco MN,

Kauffman JA, Nicolaou KC, Liu A, Brungs PH, Fusetani N (1993) Molecular basis for

the inhibition of human a-thrombin by the macrocyclic peptide cyclotheonamide-A. Proc

Natl Acad Aci USA 90:8048–8052; (c) Greco MN, Maryanoff BE (1997) In: Abell A (ed)

Advances in Amino Acid Mimetics and Peptidomimetics, vol. 1, JAI Press, Greenwich;

(d) Ganesh V, Lee AY, Clardy J, Tulinsky A (1996) Comparison of the structures of the

cyclotheonamide A complexes of human a-thrombin and bovine b-trypsin. Protein Sci

5:825–835

162. (a) Fusetani N, Nakao Y, Matsunaga S (1991) Bioactive marine metabolites. 39.

Nazumamide A, a thrombin-inhibitory Tetrapeptide, from a marine sponge, Theonella sp.

Tetrahedron Lett 32:7073–7074; (b) Hayashi K, Hamada Y, Shioiri T (1992) Synthesis of

nazumamide A, a thrombin-inhibitory linear tetrapeptide, from a marine sponge, Theonellasp. Tetrahedron Lett 33:5075–5076

163. Nienaber VL, Amparo EC (1996) A noncleavable retro-binding peptide that spans the

substrate binding cleft of serine proteases. Atomic structure of nazumamide A: human

thrombin. J Am Chem Soc 118:6807–6810

164. (a) Nakao Y, Matsunaga S, Fusetani N (1993) Toxadocial A: a novel thrombin inhibitor from

the marine sponge Toxadocia cylindrica. Tetrahedron Lett 34:1511–1514; (b) Nakao Y,

Matsunaga S, Fusetani N (1993) Toxadocial B, toxadocial C and toxadocic acid

A: thrombin-inhibitory aliphatic tetrasulfates from the marine sponge, Toxadocia cylindrica.Tetrahedron 49:11183–11188

165. (a) Carroll AR, Pierens GK, Fechner G, Leone P, Ngo A, Simpson M, Hyde E, Hooper JNA,

Bostrom SL, Musil D, Quinn RJ (2002) Dysinosin A: a novel inhibitor of factor Vila and

thrombin from a new genus and species of Australian sponge of the family Dysideidae. J Am

Chem Soc 124:13340–13341; (b) Carroll AR, Buchanan MS, Edser A, Hyde E, Simpson M,

Quinn RJ (2004) Dysinosins B-D, inhibitors of factor VIIa and thrombin from the Australian

sponge Lamellodysidea chlorea. J Nat Prod 67:1291–1294

166. (a) Murakami M, Ishida K, Okino T, Okita Y, Matsuda H, Yamaguchi K (1995) Aeruginosins

98-A and 98-B, trypsin inhibitors from the blue-green algaMicrocystis aeruginosa (NIES-98).Tetrahedron Lett 36:2785–2788; (b) Matsuda H, Okino T, Murakami M, Yamaguchi K,

(1996) Aeruginosins 102-A and B, new thrombin inhibitors from the cyanobacterium

Microcystis viridis (NIES-102). Tetrahedron 46:14501–14506; (c) Ishida K, Okita Y,

Matsuda H, Okino T, Murakami M (1999) Aeruginosins, protease inhibitors from the cyano-

bacterium Microcystis aeruginosa. Tetrahedron 55:10971–10988

167. Shin HJ, Matsuda H, Murakami M, Yamaguchi K (1997) Aeruginosins 205A and -B, serine

protease inhibitory glycopeptides from the cyanobacterium Oscillatoria agardhii (NIES-205). J Org Chem 62:1810–1813

168. (a) Hanessian S, Ersmark K, Wang X, Valle JRD, Blomberg N, Xue Y, Fjellstrom O (2007)

Structure-based organic synthesis of unusual aeruginosin hybrids as potent inhibitors of

thrombin. Bioorg Med Chem Lett 17:3480–3485; (b) Ersmark K, Valle JRD, Hanessian

S (2008) Chemistry and biology of the aeruginosin family of serine protease inhibitors.

Angew Chem Int Ed 47:1202–1223

1224 Y. Nakao and N. Fusetani

Page 81: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

169. Buchanan MS, Carroll AR, Wassling D, Jobling M, Avery VM, Davis RA, Feng Y, Xue Y,

Oster L, Fex T, Deinum J, Hooper JNA, Quinn RJ (2008) Clavatadine A, a natural products

with selective recognition and irreversible inhibition of factor Xia. J Med Chem 51:

3583–3587

170. Buchanan MS, Carroll AR, Wassling D, Jobling M, Avery VM, Davis RA, Feng Y, Hooper

JNA, Quinn RJ (2009) Clavatadines C-E, guanidine alkaloids from the Australian sponge

Suberea clavata. J Nat Prod 72:973–975

171. Fusetani N, Fujita M, Nakao Y, Matsunaga S (1999) Tokaramide A, a new cathepsin

B inhibitor from the marine sponge Theonella aff. mirabilis. Bioorg Med Chem Lett

9:3397–3402

172. (a) Nakao Y, Fujita M, Warabi K, Matsunaga S, Fusetani N (2000) Bioactive marine

metabolites. Part 104. Miraziridine A, a novel cysteine protease inhibitor from the marine

sponge Theonella aff. mirabilis. J Am Chem Soc 122:10462–10463; (b) Konno H, Kubo K,

Makabe H, Toshiro E, Hinoda N, Nosaka K, Akaji K (2007) Total synthesis of miraziridine

A and identification of its major reaction site for cathepsin B. Tetrahedron 63:9502–9513

173. Murayama S, Nakao Y, Matsunaga S (2008) Asteropterin, an inhibitor of cathepsin B, from

the marine sponge Asteropus simplex. Tetrahedron Lett 49:4186–4188

174. (a) Potts BCM, Faulkner DJ, Chan JA, Simolike GC, Offen P, Hemling ME, Francis TA

(1991) Didemnaketals A and B, HIV-1 protease inhibitors from the ascidian Didemnum sp.

J Am Chem Soc 113:6321–6322; (b) Salomon CE, Williams DH, Lobkovsky E, Clardy JC,

Faulkner DJ (2002) Relative and absolute stereochemistry of the didemnaketals, metabolites

of a Palauan ascidian, Didemnum sp. Org Lett 4:1699–1702

175. (a) Wang PZ, Tu YQ, Yang L, Dong CZ, Kitching W (1998) Synthetic studies of

didemnaketal analogue – construction of the (+)- and (�)-5,6-dihydroxy-3,7-dimethyl-

octanal intermediates. Tetrahedron Asymmetry 9:3789–3795; (b) Jia YX, Wu B, Li X,

Ren SK, Tu YQ, Chan ASC, Kitching W (2001) Synthetic studies of the HIV-1 protease

inhibitive didemnaketals: stereocontrolled synthetic approach to the key mother spiroketals.

Org Lett 3:847–849; (c) Jia YX, Li X, Wang PZ, Wu B, Zhao X, Tu Y (2002) Convergent

synthesis of the spiroketal core of the HIV-1 protease inhibitors the didemnaketals. J Chem

Soc Perkin Trans 1 565–570; (d) Jia YX, Li X, Wu B, Zhao XZ, Tu YQ (2002) A convergent

synthesis of the spiroketal moiety of the HIV-1 protease inhibitors didemnaketals.

Tetrahedron 58:1697–1708; (e) Zhao XZ, Tu YQ, Peng L, Li XQ, Jia YX (2004)

Synthetic studies of the HIV-1 protease inhibitive didemnaketals: stereocontrolled

synthesis of an ester side chain. Tetrahedron Lett 45:3713–3716; (f) Zhao XZ, Peng L,

Tang M, Tu YQ, Gao SH (2005) Synthetic studies of the HIV-1 protease inhibitive

didemnaketals: precise and stereocontrolled synthesis of the key mother spiroketal. Tetra-

hedron Lett 46:6941–6944

176. Fan X, Flentke GR, Rich DH (1998) Inhibition of HIV-1 protease by a subunit of

didemnaketal A. J Am Chem Soc 120:8893–8894

177. Fujita M, Nakao Y, Matsunaga S, Seiki M, Itoh Y, van Soest RWM, Fusetani N (2001)

Ancorinosides B-D, inhibitors of membrane type 1 matrix metalloproteinase (MT1-MMP),

from the marine sponge Penares sollasi Thiele. Tetrahedron 57:1229–1234

178. Ohta S, Ohta M, Ikegami S (1997) Ancorinoside A: a novel tetramic acid glycoside from the

marine sponge. Ancorina sp. which specifically inhibits blastulation of starfish embryos.

J Org Chem 62:6452–6453

179. Fujita M, Nakao Y, Matsunaga S, Seiki M, Itoh Y, van Soest RWM, Heubes M, Faulkner DJ,

Fusetani N (2001) Isolation and structure elucidation of two phosphorylated sterol sulfates,

MT1-MMP inhibitors from a marine sponge Cribrochalina sp.: revision of the structures of

haplosamates A and B. Tetrahedron 57:3885–3890

180. Fujita M, Nakao Y, Matsunaga S, Seiki M, Itoh Y, Yamashita J, van Soest RWM, Fusetani

N (2003) Bioactive marine metabolites, Part 124. Ageladine A: an antiangiogenic matrixme-

talloproteinase inhibitor from the marine sponge Agelas nakamurai. J Am Chem Soc

125:15700–15701

23 Enzyme Inhibitors from Marine Invertebrates 1225

Page 82: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

181. (a) Meketa ML, Weinreb SM (2006) Total synthesis of ageladine A, an angiogenesis

inhibitor from the marine sponge Agelas nakamurai. Org Lett 8:1443–1446; (b) Shengule

SR, Karuso P (2006) Concise total synthesis of the marine natural product ageladine A. Org

Lett 8:4083–4084; (c) Ando N, Terashima S (2007) Synthesis and matrix metalloproteinase

(MMP)-12 inhibitory activity of ageladine A and its analogs. Bioorg Med Chem Lett

17:4495–4499; (d) Ando N, Terashima S (2009) Synthesis of novel ageladine A analogs

showing more potent matrix metalloproteinase (MMP)-12 inhibitory activity than the natural

product. Bioorg Med Chem Lett 19:5461–5463

182. Bayle JH, Crabtree GR (1997) Protein acetylation: more than chromatin modification to

regulate transcription. Chem Biol 4:885–888

183. (a) Sambucetti LC, Fischer DD, Zabludoff S, Kwon PO, Chamberlin H, Trogani N, Xu H,

Cohen D (1999) Histone deacetylase inhibition selectively alters the activity and expression

of cell cycle proteins leading to specific chromatin acetylation and antiproliferative effects.

J Biol Chem 274:34940–34947; (b) Sato N, Ohta T, Kitagawa H, Kayahara M, Ninomiya I,

Fushida S, Fujimura T, Nishimura G, Shimizu K, Miwa K (2004) FR901228, a novel histone

deacetylase inhibitor, induces cell cycle arrest and subsequent apoptosis in refractory human

pancreatic cancer cells. Int J Oncol 24:679–685

184. (a) Minucci S, Pelicci PG (2006) Histone deacetylase inhibitors and the promise of epige-

netic (and more) treatments for cancer. Nat Rev Cancer 6:38–51; (b) Han JW, Ahn SH, Park

SH, Wang SY, Bae GU, Seo DW, Kwon HK, Hong S, Lee HY, Lee YW, Lee HW

(2000) Apicidin, a histone deacetylase inhibitor, inhibits proliferation of tumor cells via

induction of p21(WAF1/Cip1) and gelsolin. Cancer Res 60:6068–6074; (c) Ju R, Muller MT

(2003) Histone deacetylase inhibitors activate p21(WAF1) expression via ATM. Cancer Res

63:2891–2897

185. (a) Insinga A, Monestiroli S, Ronzoni S, Gelmetti V, Marchesi F, Viale A, Altucci L, Nervi

C, Minucci S, Pelicci PG (2005) Inhibitors of histone deacetylases induce tumor-selective

apoptosis through activation of the death receptor pathway. Nat Med 11:71–76; (b) Nebbioso

A, Clarke N, Voltz E, Germain E, Ambrosino C, Bontempo P, Alvarez R, Sch-avone EM,

Ferrara F, Bresciani F, Weisz A, de Lera AR, Gronemeyer H, Altucci L (2005) Tumor-

selective action of HDAC inhibitors involves TRAIL induction in acute myeloid leukemia

cells. Nat Med 11:77–84

186. (a) Munster PN, Troso-Sandoval T, Rosen N, Rifkind R, Marks PA, Richon VM (2001) The

histone deacetylase inhibitor suberoylanilide hydroxamic acid induces differentiation of

human breast cancer cells. Cancer Res 61:8492–8497; (b) Jung M, Brosch G, Kolle D,

Scherf H, Gerh€auser C, Loidl P (1999) Amide analogues of trichostatin A as inhibitors of

histone deacetylase and inducers of terminal cell differentiation. J Med Chem 42:4669–4679

187. Deroanne CF, Bonjean K, Servotte S, Devy L, Colige A, Clausse N, Blacher S, Verdin E,

Foidart JM, Nusgens BV, Castronovo V (2002) Histone deacetylases inhibitors as anti-

angiogenic agents altering vascular endothelial growth factor signaling. Oncogene 21:427–436

188. (a) Pina IC, Gautschi JT, Wang GYS, Sanders ML, Schmitz FJ, France D, Cornell-Kennon S,

Sambucetti LC, Remiszewski SW, Perez LB, Bair KW, Crews P (2003) Psammaplins from

the sponge Pseudoceratina purpurea: inhibition of both histone deacetylase and DNA

methyltransferase. J Org Chem 68:3866–3873; (b) Godert AM, Angelino N, Woloszynska-

Read A, Morey SR, James SR, Karpf AR, Sufrin JR (2006) An improved synthesis of

psammaplin A. Bioorg Med Chem Lett 16:3330–3333

189. Remiszewski SW, Sambucetti LC, Bair KW, Bontempo J, Cesarz D, Chandramouli N, Chen

R, Cheung M, Cornell-Kennon S, Dean K, Diamantidis G, France D, Green MA, Howell KL,

Kashi R, Kwon P, Lassota P, Martin MS, Mou Y, Perez LB, Sharma S, Smith T, Sorensen E,

Taplin F, Trogani N, Versace R, Walker H, Weltchek-Engler S, Wood A, Wu A, Atadja

P (2003) N-hydroxy-3-phenyl-2-propenamides as novel inhibitors of human histone

deacetylase with in vivo antitumor activity: discovery of (2E)-N-hydroxy-3-[4-[[(2-hydroxyethyl)[2-(1H-indol-3-yl)ethyl]amino]methyl]-phenyl]-2-propenamide (NVP-

LAQ824). J Med Chem 46:4609–4624

1226 Y. Nakao and N. Fusetani

Page 83: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

190. Simmons TL, Andrianasolo E, Mcphail K, Flatt P, Gerwick W (2005) Marine natural

products as anticancer drugs. Mol Cancer Ther 4:333–342

191. Oku N, Nagai K, Terada Y, van Soest RWM, Matsunaga S, Fusetani N (2004) Three new

cyclostellettamines, which inhibit histone deacetylase, from a marine sponge of the genus

Xestospongia. Bioorg Med Chem Lett 14:2617–2620

192. (a) Nakao Y, Yoshida S, Matsunaga S, Shindoh N, Terada Y, Nagai K, Yamashita JK,

Ganesan A, van Soest RWM, Fusetani N (2006) Azumamides A-E: histone deacetylase

inhibitory cyclic tetrapeptides from the marine spongeMycale izuensis. Angew Chem Int Ed

45:7553–7557; (b) Izzo I, Maulucci N, Bifulco G, De Riccardis F (2006) Total synthesis of

azumamides A and E. Angew Chem Int Ed 45:7557–7560

193. Fusetani N, Sugano M, Matsunaga S, Hashimoto K (1987) (+)-Curcuphenol and dehydrocur-

cuphenol, novel sesquiterpenes which inhibit H,K-ATPase, from a marine sponge Epipolasissp. Experientia 43:1234–1235

194. McEnroe FJ, Fenical W (1978) Structures and synthesis of some new antibacterial

sesquiterpenoids from gorgonian coral Pseudopterogorgia rigida. Tetrahedron 34:1661–

1664

195. Fusetani N, Sugano M, Matsunaga S, Hashimoto K, Shikama H, Ohta A, Nagano H (1987)

Isolation of a hexaprenylhydroquinone sulfate from the marine sponge Dysidea sp. as an H,

K-ATPase inhibitor. Experientia 43:1233–1234

196. Sata NU, Sugano M, Matsunaga S, Fusetani N (1999) Bioactive marine metabolites – Part 88

– Sinulamide: an H,K-ATPase inhibitor from a soft coral Sinularia sp. Tetrahedron Lett

40:719–722

197. (a) Nakamura H, Wu H, Ohizumi Y, Hirata Y (1984) Agelasines A, B, C and D, novel

bicyclic diterpenoids with a 9-methyladeninium unit possessing inhibitory effects on Na,K-

ATPase from the Okinawan sea sponge Agelas sp. Tetrahedron Lett 25:2989–2992; (b)

Wu H, Nakamura H, Kobayashi J, Ohizumi Y, Hirata Y (1984) Agelasines E and F, novel

monocyclic diterpenoids with a 9-methyladeninium unit possessing inhibitory effects on Na,

K-ATPase isolated from the Okinawan sea sponge Agelas nakamurai Hoshino. TetrahedronLett 25:3719–3722; (c) Wu H, Nakamura H, Kobayashi J, Kobayashi M, Ohizumi Y, Hirata

Y, (1986) Physiologically active marine natural products from porifera. 12. Structures of

agelasines, diterpenes having a 9-methyladeninium chromophore isolated from the Okina-

wan marine sponge Agelas nakamurai Hoshino. Bull Chem Soc Jpn 59:2495–2504

198. Nakamura H, Wu H, Kobayashi J, Kobayashi M, Ohizumi Y, Hirata Y (1985) Agelasidines –

novel hypotaurocyamine derivatives from the Okinawan sea sponge Agelas nakamuraiHoshino. J Org Chem 50:2494–2497

199. Okamoto Y, Ojika M, Sakagami Y (1999) Iantheran A, a dimeric polybrominated benzofuran

as a Na,K-ATPase inhibitor from a marine sponge, Ianthella sp. Tetrahedron Lett 40:507–510200. Okamoto Y, Ojika M, Kato S, Sakagami Y (2000) Ianthesines A-D, four novel

dibromotyrosine-derived metabolites from a marine sponge, Ianthella sp. Tetrahedron

56:5813–5818

201. Nakamura H, Kobayashi J, Kobayashi M, Ohizumi Y, Hirata Y (1985) Physiologically active

marine natural product from Porifera.7. Xestoquinone: a novel cardiotonic marine natural

product isolated from the Okinawan sea sponge Xestospongia sapra. Chem. Lett 713–716

202. (a) Kobayashi M, Shimizu N, Kyogoku Y, Kitagawa I (1985) Halenaquinol and halenaquinol

sulfate, pentacyclic hydroquinones from the Okinawan marine sponge Xestospongia sapra.Chem Pharm Bull 33:1305–1308; (b) Kobayashi M, Shimizu N, Kitagawa I, Kyogoku Y,

Harada N, Uda H (1985) Absolute stereostructures of halenaquinol and halenaquinol sulfate,

pentacyclic hydroquinones from the Okinawan marine sponge Xestospongia sapra, as

determined by theoretical calculation of CD spectra. Tetrahedron Lett 26:3833–3836

203. Gorshkova IA, Gorshkov BA, Fedoreev SA, Shestak OP, Novikov VL, Stonik VA

(1999) Inhibition of membrane transport ATPases by halenaquinol, a natural cardioactive

pentacyclic hydroquinone from the sponge Petrosia seriata. Comp Biochem Physiol

C 122:93–99

23 Enzyme Inhibitors from Marine Invertebrates 1227

Page 84: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

204. Stonik VA, Makarieva TN, Dmitrenok AS (1992) Sarcochromenol sulfate A,

sarcochromenol sulfate B, sarcochromenol sulfate C, and sarcohydroquinone sulfate A,

sarcohydroquinone sulfate B, sarcohydroquinone sulfate C, new natural products from the

sponge Sarcotragus spinulosus. J Nat Prod 55:1256–1260

205. Kalaitzis JA, Leone PD, Harris L, Butler MS, Ngo A, Hooper JNA, Quinn RJ

(1999) Adociasalfates 1, 7, and 8: new bioactive hexaprenoid hydroquinones from the

marine sponge Adocia sp. J Org Chem 64:5571–5574

206. (a) Erickson KL, Beutler JA, Cardelina II JH, Boyd MR (1997) Salicylihalamides A and B,

novel cytotoxic macrolides from the marine sponge Haliclona sp. J Org Chem 62:8188–

8192; (b) Erickson KL, Beutler JA, Cardellina II JH, Boyd MR (2001) Salicylihalamides

A and B, novel cytotoxic macrolides from the marine sponge Haliclona sp. J Org Chem

66:1532–1532

207. (a) Galinis DL, McKee TC, Pannell LK, Cardellina II JH, Boyd MR (1997) Lobatamides

A and B, novel cytotoxic macrolides from the tunicate Aplidium lobatum. J Org Chem

62:8968–8969; (b) McKee TC, Galinis DL, Pannell LK, Cardellina II JH, Laakso J, Ireland

CM, Murray L, Capon RJ, Boyd MR (1998) The lobatamides, novel cytotoxic macrolides

from southwestern Pacific tunicates. J Org Chem 63:7805–7810

208. Suzumura K, Takahashi I, Matsumoto H, Nagai K, Setiawan B, Rantiatmodjo RM, Suzuki K,

Nagano N (1997) Structural elucidation of YM-75518, a novel antifungal antibiotic isolated

from Pseudomonas sp. Q38009. Tetrahedron Lett 38:7573–7576

209. (a) Boyd MR, Farina C, Belfiore P, Gagliardi S, Kim JW, Hayakawa Y, Beutler JA, McKee

TC, Bowman BJ, Bowman EJ (2001) Discovery of a novel antitumor benzolactone enamide

class that selectively inhibits mammalian vacuolar-type (H+)-ATPases. J Pharmacol Exp

Ther 297:114–120; (b) Wu Y, Liao X, Wang R, Xie XS, De Brabander JK (2002) Total

synthesis and initial structure-function analysis of the potent V-ATPase inhibitors salicyli-

halamide A and related compounds. J Am Chem Soc 124:3245–3253; (c) Shen RC, Lin CT,

Bowman EJ, Bowman BJ, Porco Jr. JA (2003) Lobatamide C: total synthesis, stereochemical

assignment, preparation of simplified analogues, and V-ATPase inhibition studies. J Am

Chem Soc 125:7889–7901

210. Diyabalanage T, Amsler CD, McClintock JB, Baker BJ (2006) Palmerolide A, a cytotoxic

macrolide from the Antarctic tunicate Synoicum adareanum. J Am Chem Soc 128:5630–

5631

211. Lebar MD, Baker BJ (2007) On the stereochemistry of palmerolide A. Tetrahedron Lett

48:8009–8010

212. (a) Nicolaou KC, Guduru R, Sun YP, Banerji B, Chen DYK (2007) Total synthesis of the

originally proposed and revised structures of palmerolide A. Angew Chem Int Ed 46:5896–

5900; (b) Nicolaou KC, Sun YP, Guduru R, Banerji B, Chen DYK (2007) Total synthesis of

the originally proposed and revised structures of palmerolide A and isomers thereof. J Am

Chem Soc 130:3633–3644

213. Nicolaou KC, Leung GYC, Dethe DH, Guduru R, Sun YP, Lim CS, Chen DYK

(2007) Chemical synthesis and biological evaluation of palmerolide A analogues. J Am

Chem Soc 130:10019–10023

214. Chan GW, Francis T, Thureen DR, Offen PH, Pierce NJ, Westley JW, Johnson RK, Faulkner

DJ (1993) Purpurone, an inhibitor of ATO citrate lyase: a novel alkaloid from the marine

sponge Iotrochota sp. J Org Chem 58:2544–2546

215. Kobayashi J, Hirase T, Shigemori H, Ishibashi M, Bae MA, Tsuji T, Sasaki T (1992) New

pentacyclic compounds from the Okinawan marine sponge Xestospongia sapra. J Nat Prod55:994–998

216. Carney JR, Scheuer PJ, Kelly-Borges M (1993) Makaluvamine B, a cytotoxic pigment from

an Indonesian sponge Histodermella sp. Tetrahedron 49:8483–8486

217. Ojika M, Yoshino G, Sakagami Y (1997) Novel ceramide 1-sulfates, potent DNA topoisom-

erase I inhibitors isolated from the Bryozoa Watersipora cucullata. Tetrahedron Lett

38:4235–4238

1228 Y. Nakao and N. Fusetani

Page 85: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates

218. (a) Nemoto T, Ojika M, Sakagami Y (1997) Amphimic acids, novel unsaturated C28 fatty

acids as DNA topoisomerase I inhibitors from an Australian sponge Amphimedon sp.

Tetrahedron Lett 38:5667–5670; (b) Nemoto T, Yoshino G, Ojika M, Sakagami Y (1997)

Amphimic acids and related long-chain fatty acids as DNA topoisomerase I inhibitors from

an Australian sponge, Amphimedon sp: isolation, structure, synthesis, and biological evalu-

ation. Tetrahedron 53:16699–16710

219. Copp BR, Ireland CM (1991) Wakayin: a novel cytotoxic pyrroloiminoquinone alkaloid

from the ascidian Clavelina species. J Org Chem 56:4596–4597

220. Radisky DC, Radisky ES, Barrows LR, Copp BR, Kramer RA, Ireland CM (1993) Novel

cytotoxic topoisomerase II inhibiting pyrroloiminoquinones from Fijian sponges of the

genus Zyzzya. J Am Chem Soc 115:1632–1638

221. Venables DA, Concepcion GP, Matsumoto SS, Barrow LR, Ireland CM (1997) Makaluvamine

N: a new pyrroloiminoquinone from Zyzzya fuliginosa. J Nat Prod 60:408–410

222. Schmidt EW, Harper MK, Faulkner DJ (1995) Makaluvamines H-M and damirone C from

the Pohnpeian sponge Zyzzya fuliginosa. J Nat Prod 58:1861–1867

223. Foderaro TA, Barrows LR, Lassota P, Ireland CM (1997) Bengacarboline, a new beta-

carboline from a marine ascidian Didemnum sp. J Org Chem 62:6064–6065

224. (a) Cooray NM, Scheuer PJ, Parkanyi l, Clardy J (1988) Shermilamine A: a pentacyclic

alkaloid from a tunicate. J Org Chem 53:4619–4620; (b) Carroll AR, Cooray NM, Pointer A,

Scheuer PJ (1989) A second shermilamine alkaloid from a tunicate Trididemnum sp. J Org

Chem 54:4231–4232

225. Kobayashi J, Cheng J, Nakamura H, Ohizumi Y, Hirata Y, Sasaki T, Ohta T, Nozoe S (1988)

Ascididemin, a novel pentacyclic aromatic alkaloid with potent antileukemic activity from

the Okinawan tunicate Didemnum sp. Tetrahedron Lett 29:1177–1180

226. Schmitz FJ, DeGuzman FS, Hossain MB, van der Helm D (1991) Cytotoxic aromatic

alkaloids from the ascidian Amphicarpa meridiana and Leptoclinides sp.: Meridine and

11-hydroxyascididemin. J Org Chem 56:804–808

227. McDonald LA, Eldredge GS, Barrows LR, Ireland CM (1994) Inhibition of topoisomerase II

catalytic activity by pyridoacridine alkaloids from a Cystodytes sp. ascidian: a mechanism

for the apparent intercalator-induced inhibition of topoisomerase II. J Med Chem 37:

3819–3827

228. Carney JR, Scheuer PJ (1993) Popolohuanone E, a topoisomerase II inhibitor with selective

lung tumor cytotoxicity from the pohnpei sponge Dysidea sp. Tetrahedron Lett

34:3727–3730

229. Hamann MT, Scheuer PJ, Kelly-Borges M (1993) Biogenetically diverse, bioactive constit-

uents of a sponge, order Verongida: bromotyramines and sesquiterpene-shikimate derived

metabolites. J Org Chem 58:6565–6569

230. Juagdan EG, Kalidindi RS, Scheuer PJ, Kelly-Borges M (1995) Elenic acid, an inhibitor of

topoisomerase II, from a sponge, Plakinastrella sp. Tetrahedron Lett 36:2905–2908

231. Baz JP, Canedo LM, Tapiolas D (1996) A new tetraprenylhydroquinone derivative with an

acetic acid unit from the marine sponge Ircinia muscarum. J Nat Prod 59:960–961

232. Carney JR, Scheuer PJ (1993) A new bastadin from the sponge Psammaplysilla purpurea.J Nat Prod 56:153–157

233. Carroll AR, Feng YJ, Bowden BF, Coll JC (1996) Studies of Australian ascidians. 5.

Virenamides A-C, new cytotoxic linear peptides from the colonial didemnid ascidian

Diplosoma virens. J Org Chem 61:4059–4061

23 Enzyme Inhibitors from Marine Invertebrates 1229

Page 86: Handbook of Marine Natural Products Volume 426 || Enzyme Inhibitors from Marine Invertebrates