You are on page 1of 20

European Journal of Medicinal Chemistry 194 (2020) 112260

Contents lists available at ScienceDirect

European Journal of Medicinal Chemistry


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

Review article

Matrix metalloproteinase-9 (MMP-9) and its inhibitors in cancer: A


minireview
Subha Mondal, Nilanjan Adhikari, Suvankar Banerjee, Sk Abdul Amin, Tarun Jha*
Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, P. O. Box 17020, Jadavpur
University, Kolkata, 700032, India

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

Article history: Matrix metalloproteinases (MMPs) are zinc dependent proteolytic metalloenzyme. MMP-9 is one of the
Received 17 December 2019 most complex forms of matrix metalloproteinases. MMP-9 has the ability to degrade the extracellular
Received in revised form matrix (ECM) components and has important role in the pathophysiological functions. Overexpression
28 February 2020
and dysregulation of MMP-9 is associated with various diseases. Thus, regulation and inhibition of MMP-
Accepted 18 March 2020
Available online 21 March 2020
9 is an important therapeutic approach for combating various diseases including cancer. Inhibitors of
MMP-9 can be used as anticancer agents. Till date no selective MMP-9 inhibitors passed the clinical trials.
In this review the structure, activation, function and inhibitors of MMP-9 are mainly focused. Some
Keywords:
Matrix metalloproteinase
highly active and/or selective MMP-9 inhibitors have been discussed which may be helpful to explore the
MMP-9 structural significance of MMP-9 inhibitors. This study may be useful to design new potent and selective
MMP-9 inhibitor MMP-9 inhibitors against cancer in future.
Cancer © 2020 Elsevier Masson SAS. All rights reserved.
Hydroxamate

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2. Classification of MMPs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2.1. Collagenase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2.2. Gelatinase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2.3. Stromelysins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2.4. Matrilysins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2.5. Membrane type MMPs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2.6. Other MMPs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
3. General structure of MMPs and structural difference of MMP-9 from other MMPs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
3.1. Amino-terminal signal peptide domain . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
3.2. Pro-peptide domain (prodomain) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
3.3. Catalytic domain . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
3.4. Fibronectin domain . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
3.5. Hemopexin-like domain . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
4. Activation of MMP-9 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
5. Role of MMP-9 in cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
5.1. Metastasis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
5.2. Invasion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
5.3. Angiogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
5.4. Apoptosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
6. Role of MMP-9 in other diseases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
6.1. MMP-9 in neurological disorders . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
6.2. MMP-9 in inflammatory diseases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7

* Corresponding author.
E-mail address: tjupharm@yahoo.com (T. Jha).

https://doi.org/10.1016/j.ejmech.2020.112260
0223-5234/© 2020 Elsevier Masson SAS. All rights reserved.
2 S. Mondal et al. / European Journal of Medicinal Chemistry 194 (2020) 112260

6.3. MMP-9 in cardiovascular diseases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7


6.4. MMP-9 in lung diseases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
6.5. MMP-9 in diabetes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
7. MMP inhibitors (MMPIs) in clinical trials . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
8. MMP-9 inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
8.1. Natural MMP-9 inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
8.2. Monoclonal antibodies as MMP-9 inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
8.3. Other active MMP-9 inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
8.3.1. Hydroxamate based MMP-9 inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
8.3.2. Non hydroxamate-based MMP-9 inhibitor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
9. Trends in the development of potent and selective MMP-9 inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
10. Conclusion and future perspective . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
Declaration of interest statement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
Acknowledgement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17

1. Introduction cytokine (interleukin-1 and -6), growth factors (transforming


growth factor (TGF), tumor necrosis factor a (TNFa), platelet-
Cancer is an abnormal growth of cells and is a major group of derived growth factor (PDGF), basic fibroblast growth factor
diseases by which various parts of the body have been affected and (bFGF) and some hormones [5]. They have important roles in
lead to death worldwide. Cancer is the major cause of death after various physiological processes including development, wound
heart disease globally. In 2018, the number of death due to cancer healing, remodelling of tissues, organ morphogenesis, angiogen-
was 9.6 million [1]. There are more than 200 type of caners such as esis, etc. Angiogenesis is essential for tumor growth and develop-
lung, skin, breast, prostate, colorectal, stomach cancers, etc. Most ment during which new blood vessel are formed from the existing
common causes of cancer are high body mass index, tobacco and blood vessel [6]. In normal physiological condition, MMPs are
alcohol use, lack of physical activity, low fruit and vegetable intake, tightly regulated and expressed at low level. Dysregulation and
etc. Tobacco use (approximately 22%) and hepatitis and human overexpression of these enzymes related to various diseases
papiloma virus (HPV) (up to 25%) are liable for cancer deaths. The including neurodegenerative diseases, cardiovascular diseases,
most commonly occurred cancers and deaths caused by different lung diseases, arthritis, central nervous system (CNS) disorders
cancers are depicted in Fig. 1(A) and Fig. 1(B), respectively [1]. including epilepsy, various type of cancers, etc [4,7,8].
Cancer research mainly focused on the overexpressed molecules Therefore, MMPs have been expressed as potential diagnostic
those are associated with the survival and proliferation of cancer and are prognostic biomarker in several type of cancer [9]. MMPs
cell [1]. are regulated by various ways such as maintenance of activation of
Matrix metalloproteinases (MMPs) are zinc (Zn2þ) dependent pro-enzyme and transcription. Inhibition of MMPs function is done
endopeptidases present intracellularly and membrane bound. They by tissue inhibitor of metalloproteinase (TIMP) [3]. TIMPs are
cause deterioration of extra cellular matrix (ECM) proteins for divided into four groups TIMP-1,-2,-3 and -4. Imbalance between
example collagen, laminin, elastin, fibronectin, etc and help in the activation and inhibition of MMPs play a crucial role in patho-
extracellular matrix remodelling in various physiological and physiology of cancer [3,10].
pathological processes [2]. They can also deteriorate the non-ECM About 26 MMPs have been identified till now and most of these
molecules most of those are bioactive molecules. Another name of enzymes are present in the human proteome [11,12]. The first MMP
MMPs is matrixin those are of metzincin superfamily. They act was discovered in 1962 that is collagenase-1 (MMP-1). It is found in
depending on the metal ion such as zinc (Zn2þ) and calcium (Ca2þ) tadpole tail of North American frog species Rana catesbiana by
[3]. MMPs were first found in vertebrates and also found in in- Gross and Lapiere [5]. MMPs are secreted as zymogen (pro-
vertebrates (soyabeans, sea urchins, Arabidopsis thaliana and Cae- enzyme). They remain in inactive state because of the interaction
norrhabdita elegans) [4]. They are active at neutral pH [3]. between zinc (Zn2þ) ion of the catalytic domain with the cysteine
Expression of MMP is increased by several factors for example sulphydryl group of the pro-domain. They become activated after

Fig. 1. (A) The number of different cancers occurred in 2018. (B) The number of deaths in 2018 caused by different cancers.
S. Mondal et al. / European Journal of Medicinal Chemistry 194 (2020) 112260 3

removal of this interaction or by proteinases [13]. Depending on the serpins. MMP-11 is also called furin-activatable MMP due to its
substrate specificity MMPs are divided into six classes such as additional structural feature [8]. MMP-27 has been identified first
collagenase, gelatinase, matrilysins, stromelysins, membrane type in chicken embryo having capability to degrade gelatine and
MMPs and other type MMPs [7]. casein. It is highly expressed in human B-lymphocytes [20].
MMP-9 is the one of the most complex MMPs which belongs to
gelatinase family [14]. It inhibits or stimulates the process of 2.4. Matrilysins
degradation of ECM. It causes degradation of gelatine and type IV,
V, XI and XVI collagens during tissue remodelling which is essential Matrilysins including MMP-7 (matrilysin-1) and MMP-26
for tumour invasion and metastasis [15]. MMP-9 is usually located (matrilysin-2) do not consist of hemopexin domain. MMP-7 is
at hippocampus, cerebellum and cerebral cortex [14]. It was synthesized by epithelial cells [8]. Matrilysins are released under
discovered in 1974 and is also known as gelatinase B of proteolytic normal and physiological condition involving in the progression of
family. MMP-9 is secreted as zymogens or as the inactive form from various cancers. These cause the degradation of ECM molecule
endothelial cell, leukocyte, fibroblast, neutrophil and macrophage. (laminin, type IV collagen and entactin) and also non-ECM mole-
At the time of granulocyte differentiation, MMP-9 synthesis is cules. MMP-26 is stored intracellularly and also activates pro-
generally occurred in the bone marrow [16]. MMP9 [20]. Matrilysins help in the remodelling of postpartum
Matrix metalloproteinase inhibitors (MMPIs) act by binding to uterus and also in embryo implantation [20].
the zinc (Zn2þ) ion at the catalytic site and inhibit MMPs activity
which may be used as anticancer agents [11]. Few researchers 2.5. Membrane type MMPs
claimed that MMPIs act by inhibiting cancer cell growth whereas
some studies reported that MMPIs acts by decreasing tumour Membrane type MMPs (MT-MMPs) comprises transmembrane
proliferation by inducing apoptosis via releasing ligand [TNFa, domain or a glycosylphosphatidyl inositol anchor [8]. They are
TRAIL (Tumour necrosis factor related apoptosis inducing ligand)] divided into six subtypes such as MMP-14(MT1-MMP), MMP-
from their membrane bound inactive form [17]. 15(MT2-MMP), MMP-16 (MT3-MMP), MMP-17(MT4-MMP), MMP-
Here, we mainly focus on MMP-9 enzyme through the light of its 24 (MT5-MMP) and MMP-25(MT6-MMP) [23e25]. Among these
structure, function and inhibitors. Overexpression of MMP-9 is MT1, MT2, MT3 and MT5 have transmembrane domain. On other
associated with the various cancers such as breast, lung, prostate, hand MT4 and MT6 have glycophosphatidyl inositol anchor. MT1-
colon, gastric and pancreatic cancers are highlighted in details. MMP causes degradation of types I, II and III collagens. Other MT-
MMPs are essential for activation of pro-MMP2 and also cause
2. Classification of MMPs degradation of laminin and fibronectin [19]. It has been found that
MT1-MMP, MT2-MMP and MT3-MMP increase the invasion ca-
Depending on the substrate specificity MMPs have been classi- pacity of cancer cell by degrading fibrin [22].
fied into six major groups (Table-1). These are collagenase, gelati-
nase, stromelysin, matrilysin, membrane type and other type 2.6. Other MMPs
MMPs [7,16,18].
MMP-12, -19, 20, 21, 27, 28 are divergence in sequence
2.1. Collagenase and substrate specificity, so they cannot be properly classified in
subgroups [11]. MMP-12 causes degradation of type-IV collagen,
Collagenases including MMP-1, MMP-8, MMP-13 and MMP-18 type-I gelatine, elastin, myelin basic protein and associated with
(identified in Xenopus) are essential for cleavage of several types various pathological condition such as inflammation [22]. It also
of interstitial collagens (collagen type I, II and III) at the particular helps in the migration of microphages. It is secreted from osteo-
site to form 3/4 and 1/4 fragments [19,20]. They can also cleave ECM clasts and hypertrophic chondrocytes and expressed by macro-
molecule and non-ECM molecules. For collagenolytic activity of phages [8]. MMP-19 cleaves some ECM molecules mainly the
these MMPs, there should be a correlation between hemopexin basement membrane components. MMP-19 is also known as RASI
domain and catalytic domain [20]. (rheumatoid arthritis synovial inflammation) because it was first
identified in the active lymphocytes and rheumatoid arthritis
2.2. Gelatinase patient’s plasma [8]. MMP-20 also known as enamelysin which is
first identified in odontoblusts and also tooth specific. MMP-20 is
Gelatinases (MMP-2 and MMP-9) are liable for deterioration of an important factor for dental enamel formation. MMP-21 is
gelatine by their three fibronectin repeats and also degrade ECM identified in xenopus, found in mice and human. It cleaves gela-
like collagen (type I, IV, V, VII, X, IX and XI), laminin, elastin, fibrillin, tine but how it acts on the ECM component is not known. MMP-28
aggrecan, proteoglycans, vitronectin, etc [20,21]. MMP-9 is is also known as epilysin which is found in various tissues, for
expressed by these cells including alveolar macrophages, osteo- example, lung, heart, testis, placenta and gastrointestinal tract. It
clasts and polymorphonuclear leukocyte, etc cells [20]. It also helps in wound healing. The level of MMP-28 is increased in pa-
causes the degradation of non ECM molecules such as interleukin- tient with osteoarthritis and rheumatoid arthritis [20].
1b and TNFa. These MMPs are found to express by chondrocytes,
osteoclasts, osteoblasts, endothelial and malignant cells [22]. 3. General structure of MMPs and structural difference of
MMP-9 from other MMPs
2.3. Stromelysins
All MMPs are consisting of at least three main domains: (a)
Stromelysins including MMP-3, MMP-10 and MMP-11 are amino-terminal signal peptide domain, (b) propeptide domain and
same as collagenases but differ in that these do not cause cleavage (c) catalytic domain (Fig. 2).
of interstitial collagen. Among these MMP-3 and MMP-10 cleave
some ECM molecules and also help in the activation of pro-MMP 3.1. Amino-terminal signal peptide domain
by removing the propeptide domain where MMP-11 is less
active towards the ECM molecules but more active towards the Amino-terminal signal peptide (composed of 17e29 amino
4 S. Mondal et al. / European Journal of Medicinal Chemistry 194 (2020) 112260

Table: 1
Classification of Matrix metalloproteinases (MMPs).

Type of Sub-group Isoform Substrate Biological effect


MMP

Collagenases Collagenase-1/ MMP-1 Fibrillar and non-fibrillar collagens (Type I, II, III, Cellular proliferation and migration, increases the bioavailability of insulin like
Interstitial VI,VII,VIII and X), laminin, casein, serpins and growth factor-1 (IGF-1), keratinocyte migration, re-epithelialisation, pro-
collagenase MMP-1, -2, -9 inflammatory effects, degradation of physical barriers and proteolytic activity
in cancer progression.
Collagense-2/ MMP-8 Fibrillar collagens (Type I, II, III, V, VII, VIII and X), The activation of osteoclasts, the enhancement of collagen affinity, release of
Neutrophil gelatin, fibronectin bFGF (basic fibroblast growth factor). Regulation of mobilization and cleavage
collagenase of the a- and b-chemokines in cancer progression, anti-inflammatory effects.
Collagenase-3 MMP- Fibrillar collagens (Type I, II and III), gelatins During cancer progression- induction of EMT (epithelial to mesenchymal
13 transition), cell migration
Collagenase-4 MMP- ND e
18
Gelatinases Gelatinase A MMP-2 Basement membrane and non-fibrillar collagens Cellular migration, elevation of collagen affinity, epithelial cell migration,
(72 kDa) (Type IV, V, VII and X), fibronectin, elastin enhancement of TGF-b (transforming growth factor-b) bioavailability,
neurodegeneration through neuronal apoptosis, growth of axons,
mesenchymal cell differentiation with inflammation phenotypes, anti-
inflammatory effects, proliferation and cleaving of IGF-1 binding proteins in
progression of cancer.
Gelatinase B MMP-9 Basement membrane collagens and non fibrillar Anti-inflammatory and pro-inflammatory activity, increase in collagen
(92 kDa) collagen (Type IV, V, VII, X and XIV), gelatins, affinity, reduction of IL-2 response. Activation of TGF-b in cancer progression.
fibrillin Apoptosis of hypertrophic chondrocytes and the inclusion of newer functional
osteoblast units, resistance of tumor cell.
Matrilysins Matrilysin- MMP-7 Collagen IV-X, fibronectin, gelatins, Enhancement of TGF-b and IGF-1 bioavailability, increase in collagen affinity,
1(PUMP) proteoglycans, pro-MMP-9, laminin induction of apoptosis through activation of Fas receptor, increase of cellular
invasiveness and aggregation of abnormal cells, differentiation of adipocyte,
cellular growth and vasoconstriction, osteoclasts activation through pro-
inflammation.
Matrilysin-2/ MMP- Collagen IV, fibronectin, fibrinogen, gelatine, pro- e
endometase 26 MMP-9
Stromelysins Stromelysin-1 MMP-3 Collagens (Type III, IV, V and IX), fibronectin, Conversion of epithelialemesenchymal, generation of angiostatin-like
proteoglycan, pro-MMP-1, laminin, gelatins substances, increase in collagen affinity, bFGF release, cellular proliferation
and migration, epithelial bubble formation, bioavailability enhancement of
IGF-1 and TGF-b, pro- and anti-inflammatory effects, increase in cellular
invasiveness, apoptosis of epithelial cells, disorganization of cell aggregation,
angiogenesis upregulation, perlecan and COL-IV degradation, release of bFGF
and VEGF in cancer progression.
Stromelysin-2 MMP- Fibronectins, collagens (Type III and IV), pro- Tumstatin, angiostatin, endostatin and endorepellin generation in cancer
10 MMP-1 progression, degradation of perlecan, COL-IV and COL-XVIII in cancer
progression.
Stromrlysin-3 MMP- Serpin, aggrecan, laminin, fibronectin, gelatin Reduction of cancer cell sensitivity to NK cells and a1-proteinase inhibitor
11 release in cancer progression
Membrane MT1-MMP MMP- Gelatin, collagen (i-iii), fibronectin, casein, Cellular migration, anti-inflammatory activity, renal tubule formation, cell
type 14 laminin, MMP-2 and -13, entactin, vitronectin, reduction flattening, reduction of adhesion, transports embryo to uterine
proteoglycan. epithelium.
MT2-MMP MMP- Pro-MMP-2, gelatine, fibronectin, tenascin, Adhesion and cell flattering reduction
15 nidogen, laminin, aggrecan, perlecan, MMP-2
MT3-MMP MMP- Collagen-III, gelatin, casein, fibronectin and MMP- Adhesion and cell flattering reduction
16 2.
MT4-MMP MMP- ND e
17
MT5-MMP MMP- Fibronectin, Pro-MMP-9, pro-MMP-2, gelatine e
24
MT6-MMP MMP- Fibrin, gelatine, collagen IV, fibronectin e
25
Others Macrophage MMP- Casein, collagen-IV, vitronectin, elastin, gelatin, Causes plasminogen digestion and angiostatin release leading to the cancer
metallo elastase 12 fibronectin, fibrin fibrinogen and plasminogen. cell apoptosis, produce endostatin which form a complex with pro-MMP-9
and -13 and block their activation
RASI-1 MMP- Type-I collagen Activation of VEGF, epithelial cell migration
19
Enamelysin MMP- Gelatin, COMP, amelogenin (dentine), aggrecan, e
20
XMMP from MMP- Not determined e
Xenopus 21
CMMP from MMP- Casein, gelatin e
chicken 22
CA-MMP MMP- ND e
(cysteine array) 23
e MMP- ND e
22
Epilysin MMP- Casein Activate TGF-b proteolytically which induces epithelial to mesenchymal
28 transition in which increased invasion, migration and metastasis
Unnamed MMP- ND e
29

ND: Not determined.


S. Mondal et al. / European Journal of Medicinal Chemistry 194 (2020) 112260 5

ordinationg to the catalytic zinc in the catalytic cleft [32]. Cata-


lytic domain also contains a conserved methionine residue which
forms a ‘Met-turn’.
The catalytic cleft has six binding pockets including S1, S2, S3,
S1’, S2’ and S3’ sites. S1, S2 and S3 pocket are present at the left
handed side of the catalytic Zn2þ ion where as S1, S2´ and S3
pockets are located at the right handed side of this Zn2þ ion
expressed as primed pockets [18]. S2 and S3 pocket are solvent
expounded. Among these pockets, depth and amino acid sequence
of S1 pocket vary in different MMPs. The substrate specificity is
depending on the S1’ pocket. MMP-1 and MMP-7 have shallow S1
pocket; MMP-2, MMP-8, MMP-9, MMP-12 and MMP-14 have in-
termediate S1 pocket; and MMP-3, MMP-10, MMP-13 have deep S1
pocket [19,33]. Matrix metalloproteinase inhibitors (MMPIs) may
be designed depending on the depth and length of the S1 pocket
and also based on the amino acid sequence around this pocket
[34,35]. The S1 pocket of both MMP-2 and MMP-9 is situated in the
catalytic domain and is similar in size as well as exposed to the
solvent but differ in the residue 425e431 which form a loop in
MMP-9 that is not present in MMP-2 [18].

3.4. Fibronectin domain

Fibronectin domain consists of three repeats of fibronectin type


Fig. 2. Graphical representation of different structural domains of the MMP family II motif which are inserted into the active site domain and zinc
members.
binding domain (metalloproteinase domain) [8,36,37]. The fibro-
nectin type-II motif has the capability to bind gelatine, laminin,
acids) is responsible for its excretion out of the cell [3]. Most of collagens type-I and IV [29]. MMP-2 and MMP-9 contain fibro-
these MMPs are bound to the cell surface by a transmembrane nectin domain which is not present in other MMPs (Fig. 2). In case
domain (except MT-MMPs). of MMP-2 and -9, this fibronectin domain is a modulator
́ of collageń
recognition [18].

3.2. Pro-peptide domain (prodomain) 3.5. Hemopexin-like domain


́
The pro-peptide domain (composed of 77e87 amino acids) is The C-terminal Hemopexin domain contains about 210 aminó
responsible for activation of the enzyme. The amino acid sequence acid residues [31]. This domain is an ellipsoidal disk shaped in
of pro-domain of all MMPs is PRCGXPD that is known as “cysteine nature containing four bladed b-propellers connected with singlé
́
switch” (except MMP-23) [3,26,27]. The cysteine residue contains disulphide bond between the 1st and the 4th blades [8]. Each blade
sulfhydryl group which is coordinated to the catalytic divalent zinc consists of four antiparallel b-strands and a a-helix [31]. Generally,
́
ion to regulate the enzyme dormancy [9,26]. This “cysteine switch a calcium and a chloride ions are present at the centre of the pro-
mechanism” is pivotal [28,29]. Due to this zinc-cysteine co-ordi- peller [8]. Thiś domain can interact with substrate such as gelatin
nation, the activities of MMPs are suppressed by preventing water and collagen. In case of MMP-9, it plays an important role to bind
molecule from binding to the zinc ion which is essential for the the tissue inhibitor of metalloproteinases [9]. MMP-9 differs from
catalysis [8]. The pro-domain contains three a chains which are MMP-7 (matrilysin 1), MMP-26 (matrilysin 2) and MMP-23 because
connected with a flexible loop. These are responsible for the for- hemopexin domain is not present in these MMPs.
mation of hydrophobic pocket by interacting with each other [29]. Gelatinases (MMP-2 and MMP-9) contain an additional fibro-
nectin domain. MMP-2 and MMP-9 have similar substrate selec-
3.3. Catalytic domain tivity but differs in tissue specificity. MMP-2 and MMP-9 are highly
similar in their structure but differences are observed in the S1
On the other hand, the catalytic domain (contains 170 amino pocket. Amino acids residues 425e431 form a loop in the S1’ site of
acids approximately) is responsible for the proteolytic activity of MMP-9 which is found absent in MMP-2 [11,18]. Gelatinase B
the enzyme [30,31]. Most importantly, this domain contains a zinc- (MMP-9) is one of the most complex matrix metalloproteinase
binding consensus sequence (HEXXHXXGXXH) which is pivotal for structurally which contains the linker peptide of variable length
the proteolytic activity [24]. Catalytic domain of MMPs is struc- called hinge region (Fig. 2). Due to its complex structure, it can bind
turally spherical with a diameter of about 40 Å [29]. This domain is with various substrates such as gelatine, collagens type I and IV,
pivotal for the substrate hydrolysis [30]. Two zinc ions (one cata- procollagen type II, laminin, tissue inhibitor of metalloproteinases
lytic Zn2þ ion and one structural Zn2þ ion) are present in this (TIMPs), chemokines, etc [14,25].
domain which is required for the catalytic activity and structural
integrity respectively [8]. Five calcium ions required for the enzyme 4. Activation of MMP-9
stability and integrity [3,32].
There is a shallow catalytic cleft which divides this domain into MMPs are generally produced as pre-pro-enzyme [38,39] from
two parts-one is the N-terminal sub-domain and another is the C- which the amino-terminal signal peptide is cleaved during trans-
terminal sub-domain [32]. These two sub-domains are linked with lation and results in pro-MMPs. In order to keep the proMMPs
the highly opened U loop. Three histidine amino acid residues (i.e., inactive, the amino acid cysteine from the PRCGXPD ‘cysteine
His218, His222, His228) and a water molecule are required for co- switch’ motif coordinates with the catalytic divalent ions. Later the
6 S. Mondal et al. / European Journal of Medicinal Chemistry 194 (2020) 112260

cysteine switch is cleaved or the pro-domain is detached often by [36]. It has been found that ECM has important role in cancer
proteolytic enzymes such as the endopeptidase furin, plasmin, progression [44]. Interaction between these cells and ECM
serine proteases, or other MMPs and subsequently, results in the component is essential for cell transformation as well as carcino-
active MMP forms. In the activation of the pro-enzyme to the active genesis [5]. MMPs are highly expressed in human cancer and are
form, various activators are involved. Several pro-MMPs are acti- related to every stage of cancer development [11,36]. Cancer cell
vated by mercurial compounds, chaotropic and thiol agents. Pro- secrets various agents such as interferon, interleukins and growth
MMPs are also activated by oxidant like HOCl, ONOO. These oxi- factors as well as extracellular MMP inductor which stimulate the
dants interact with cysteine residue in the pro-peptide domain surrounding host cell to generate required MMPs for the tumor cell
under inflammatory condition. On the other hand, the endogenous [5].
TIMPs counteract these actions. Early MMPs expressions in tumor cells help in the remodelling
Similarly, the MMP-9 enzyme is secreted as pro-MMP-9 which is of ECM and release membrane-bound growth factors to lay out
an inactive form of MMP-9. The activation of pro-MMP-9 (molec- microenvironment for the tumorigenesis [36]. The role of MMP-9 in
ular weight 92 kDa) by various MMP-9 activators occurs in two angiogenesis, metastasis and cancer invasion has been identified
steps (Fig. 3). (Fig. 4). MMP-9 is also responsible for survival and spreading of
Each step decreased molecular weight (MW) by approximately cancer cells.
5 kDa. In the first step, a cleavage at glutamate-59 is occurs to Cancer cells in a paracrine manner, by secreting interleukin,
produce an intermediate from (MW: 86 kDa). The second step deals interferon, growth factors and extracellular MMP inductors, stim-
with a cleavage at arginine-106 to produce active MMP-9 (MW: ulate surrounding host cells to produce required MMPs. MMPs
82 kDa) [40,41]. Post-activation fate of MMP-9 results in the secreted by normal cells can be bounded on the cancer cell surface
cleavage of the C-terminal hemopexin-like domain (MW: 65 kDa and used by the tumor cells. They are involved in all steps during
species) or a direct removal of the catalytic site leading to an carcinogenesis [15].
inactive from (MW: 50e60 kDa) [42]. MMP-9 is found to play a crucial role in gastric cancer pro-
The enzyme proteolysis of the prodomain is an interesting gression as detected by SDS-PAGE and SDS-PAGE zymography as-
mechanism of MMP-9 activation. MMP-9 activators are MMP-2, -3, says [45]. MMP-9 gene polymorphism has important roles in breast
-7, -10, 13, cathepsin G and plasmin [21,22]. Interestingly, the cancer patients along with identification of risk of developing
MMP-3 is the most potent activator of MMP-9 [42]. It has been breast cancer [46]. It is noticed that MMP-9 has a direct correlation
found that thrombospondin-1 also increases the activation of of poor prognosis in breast tumors [47].
MMP-9 [43]. It has been found that the avb6 integrin is expressed in colon
cancers which in turn triggers MMP-9 secretion and subsequently,
5. Role of MMP-9 in cancer involves the protein-kinase-C pathway activation [48]. Further
study showed that avb6-mediated secretion of MMP-9 has a crucial
Cancer research is mainly focused on the overexpressed mole- role in colon cancer progression [49]. MMP-9 overexpression is
cules and the process by which these help in cancer cell progression associated with decreased survival rate and metastasis in breast as
well as colon cancers [50].
MMP-9 is also found to be induced by vascular endothelial
growth factor (VEGF) during lung metastasis and subsequently,
deficiency of MMP-9 helps to reduce the metastasis [51]. Also,
MMP-9 has crucial roles in several cancers associated with
inflammation [50]. Gelatinases mainly MMP-9 is related to the
negative regulation of immune response to cancer by cleaving
interleukin-2a (IL-2a) as well as through shedding of intercellular

Fig. 3. Schematic representation of the activation process of MMP-9. Fig. 4. Role of MMP-9 in cancer progression.
S. Mondal et al. / European Journal of Medicinal Chemistry 194 (2020) 112260 7

adhesion molecule-1 (ICAM-1) and transforming growth factor diseases and cardiovascular to lung diseases [55]. The correlation of
(TGF-b) activation [50]. MMP-9 with different diseases and disorders are given in Fig. 5.

5.1. Metastasis 6.1. MMP-9 in neurological disorders

Metastasis is the main culprit of death in cancer patients. MMP- MMP-9 plays important roles in pathophysiological mechanism
9 causes degradation of gelatin and type IV, V, XI and XVI collagens of various neurological and neurodegenerative diseases such as
which are essential for metastasis [14]. MMP-9 promotes metas- Alzheimer’s disease (AD) and Parkinson’s disease (PD) through
tasis through breakdown of physical barrier of ECM. Metastasis remodelling and degradation of ECM, blood-brain barrier (BBB)
occurs in several steps. Initially there is a disconnection from disruption, inflammation and microglial activation [56].
intracellular part and releasing of a tumour cell. After that degra- MMP-9 has crucial roles in neurological disorders like stroke
dation of ECM component occurs and migration of the cell takes (Fig. 5). Gu et al. [57] reported that during cerebral ischemia MMP-9
place. Then the cell penetrates into the blood and lymphatic vessel become activated through oxidation and S-nitrosylation leading to
and adhered to the endothelial cell. Finally leads toward the growth the cortical neuronal apoptosis. MMP-9 is also associated with
of a secondary tumour cell in the other part of the body. Itoh and pathogenesis of epilepsy. MMP-9 activity also can impair synaptic
co-workers [52] showed a prime contribution of host-derived plasticity and mossy fibre sprouting within hippocampus, thus
MMP-9 in tumor metastasis. Thereby, the MMP-9 activity leading to the formation of epileptic foci. It has important role in
reduced by inhibitors is useful in the metastasis. pathology of memory learning, inflammatory diseases, psychiatric
diseases (schizophrenia, bipolar disorder), addiction and multiple
5.2. Invasion sclerosis [14].

MMP-9 causes the degradation of type IV collagen of basal 6.2. MMP-9 in inflammatory diseases
membrane near the tumor cell that invades the other tissue, and
therefore induces metastasis [12]. DNA repair protein Ku interact MMP-9 is associated with acute and chronic inflammatory dis-
with the pro-MMP-9 (MW: 85 kDa) at the cell surface of leukemia eases (Fig. 5) [41]. It has both pro and anti-inflammatory effects.
cell lines to form Ku/MMP-9 complex which regulates the cell in- MMP-9 enables influx of leukocyte in inflammation and affects
vasion [53,54]. Therefore, by blocking the MMP-9 there may be an permeability of blood brain barrier, thus promotes inflammation.
inhibition of the Ku/MMP-9 expression mediated cancer cell inva- MMP-9 inhibitors may be useful in organ specific autoimmune
sion on the cell surface [53]. diseases such as rheumatoid arthritis [41].

5.3. Angiogenesis 6.3. MMP-9 in cardiovascular diseases

This process is essential for growth and development of tumor MMP-9 gene polymorphism is associated with cardiovascular
cell. Some ECM components such as fibronectin, thrombospondin- diseases including hypertension, atherosclerosis and myocardial
1, laminin and osteopontin affect the phenotype of tumor by infarction [16]. MMP-9 causes proteolytic degradation of protein
modulationg cancer cell migration and angiogenesis [5]. MMPs and release vascular endothelial growth factor through which
endorse angiogenesis by degrading the basement membrane and MMP-9 plays a critical role in neovascularisation.
ECM component. Due to disruption of basement membrane, the
endothelial cell migrates from the existing vessel to produce new 6.4. MMP-9 in lung diseases
blood vessel and also releases ECM bound factor [5]. The activity of
MMP causes the uncovering of hidden epitopes of ECM proteins. Normal function in lung requires alveolar support by ECM.
MMP-9 digests type IV collagen revealing at the same time HUIV26- Therefore, excess degradation and abnormal remodelling of ECM
epitope and regulating blood vessels development. MMP-9 is
involved in the mechanism of angiogenesis (Fig. 4).
However, MMPs may inhibit the mechanism of angiogenesis. It
causes the degradation of plasminogen to release angiostatin and
also cleaves the collagen XVIII to produce endostatin and thereby
inhibits the process of angiogenesis. MMP-2, -7, -9 and 12 help in
the digestion of plasminogen to release angiostatin. This resulting
angiostatin can increase an apoptosis in tumor cells [5].

5.4. Apoptosis

MMPs have both pro-apoptotic and anti-apoptotic activity. The


pro-apoptotic activity is related to the change in ECM composition.
The anti-apoptotic activities are cleavage of Fas ligand, activation of
serine or protein kinase B or threonine kinase AKT [5]. MMPs cleave
the adhesion molecule and lead to apoptosis. Angiostatin causes
apoptosis in cancer cells and endostatin binds with the pro-MMP9
to produce a stable complex that inhibits MMP-9 activation.

6. Role of MMP-9 in other diseases

The MMP-9 gene polymorphism 1562C/T brought about


interesting result from cancer to neurological/neurodegenerative Fig. 5. Schematic representation of diseases associated with the MMP-9.
8 S. Mondal et al. / European Journal of Medicinal Chemistry 194 (2020) 112260

causes many respiratory disorders such as chronic obstructive hydroxamate MMPI (Fig. 6). It inhibited major MMPs (such as
pulmonary diseases (COPD), bronchial asthma and sometime MMP-1, -2, -3, -7 and -9). It was found to be orally bioavailable
leading to tuberculosis (Fig. 5). Numerous cells present in lung such (20e50%) due to the presence of a-hydroxyl and tert-butyl groups
as epithelium, fibroblast, macrophages and myofibroblasts have those increased aqueous solubility [65]. This compound is effective
ability to express MMPs those are associated with early stage of against breast and lung metastasis models in pre-clinical studies
chronic obstructive pulmonary disease (COPD) [58]. Increased [73]. However, it has some major adverse effects including toxicity
MMP-9 activity is found in various lung diseases. MMP-9 in COPD of gastro-intestinal tract, weight loss, inflammation, fibrosis, hae-
alters ECM to enlarge the air space in lungs. It is reported that a morrhage and necrosis of tissues [73]. Phase II study of marimastat
selective inhibitor of MMP-9 and -12 decreases the inflammatory with patients having different types of cancers resulted in poor
process correlated with exposure to cigarette smoke [59]. Treat- patient compliance and only 5% patients responded to the therapy
ment with MMP-9 inhibitor is also protective in six-month smoke [74]. However, marimastat entered into phase III clinical studies
exposure patient. with patients having various solid tumors in combination chemo-
therapy. It showed a significant improvement for patients having
6.5. MMP-9 in diabetes advanced gastric cancer [74].
CGS-27023A (3), a sulphonamide derivative is also known as
The consequence of hyperglycemia on MMPs regulation in MMI-270 (Fig. 6). It is a broad-spectrum non-peptidomimetic
vascular cells is little known [60]. Uemura and co-workers [61] MMPI that also inhibits tumor growth and angiogenesis as evi-
found an increased MMP-9 expression in plasma and vascular tis- denced in preclinical studies [75]. It is a broad spectrum MMPI and
sue of diabetic animal models. It is also reported that oxidative orally available. It acts as an inhibitor against MMP-1
stress is a key factor in glucose-induced MMP-9 activity. Ebihara (IC50 ¼ 33 nM), MMP-2 (IC50 ¼ 11 nM), MMP-3 (IC50 ¼ 13 nM),
et al. [62] suggested that increased plasma MMP-9 concentrations MMP-9 (IC50 ¼ 8 nM) and MMP-13 (IC50 ¼ 6 nM) [66]. However,
lead up to the microalbuminuria development in non-insulin- this compound was failed in clinical trial because of its side effect
dependent diabetes mellitus (NIDDM) patients. Nguyen and col- including joint and muscle pain [29].
leagues [63] explored roles of MMP-8 and -9 in diabetic wound GM6001 (4) is another broad spectrum, potent and non-
healing (Fig. 5). selective MMPI (Fig. 6). It blocks both MMP-3 and MMP-9 enzy-
matic activity [65]. It is a hydroxamic acid derivative and the
7. MMP inhibitors (MMPIs) in clinical trials hydroxamic acid moiety chelate with the Zn2þ ion to form biden-
tate complex.
MMPs not only have crucial roles in cell invasion, angiogenesis GI129471 (5) is a broad spectrum synthetic MMPI can upregu-
and metastasis but also have important roles in cell transformation, lated both synthesis and transcription of pro-MMP-9. It can inhibit
signal transduction, apoptosis, cell growth and immune regulation. human fibrosarcoma HT1080 invasion through a reduced proteo-
Therefore, MMPIs are required for treatment of such conditions lytic activity [68].
[37]. Depending on the presence of the zinc ion and substrate CGS-25966 (6) is a sulphonamide based hydroxamic acid de-
binding pocket, various synthetic MMPIs have been designed [18]. rivative and close analog of CGS-27023A [65,69]. Replacement of
Zinc binding groups (ZBGs) including hydroxamic acid, thiols, car- the 4-pyridylmethyl moiety of CGS-27023A (3) with a benzyl group
boxylates and phosphonic acid are identified [64,65]. Among these results in compound CGS-25966 (Fig. 6). The (R)-enantiomer of this
groups, hydroxamic acid is more preferred because of its Zn2þ compound showed potent MMP inhibition (IC50 ¼ 27 nM for MMP-
binding ability. Due to the presence of hydrogen bond between the 9 and IC50 ¼ 11 nM for MMP-2) while (S)-enantiomer showed low
heteroatom of ZBG and neighbouring amino acids at MMP active or no inhibition [69].
site, the NH and deprotonated OH groups of hydroxamic acid form However, the above mentioned broad spectrum and non-
hydrogen bond with Ala and Glu residues [18,36]. selective MMPIs were failed in clinical trials due to their poor sol-
The first generation of MMPIs are basically peptidomimetic ubility, low oral bioavailability and severe side effects including
compounds having hydroxamic acid moiety. Batimastat/BB-94 (1) musculoskeletal syndrome (join stiffness, pain, tendinitis and
(IC50 ¼ 1 nM for MMP-9 and IC50 ¼ 4 nM for MMP-2) and inflammation) [13,18]. The other disadvantages of these com-
Marimastat/BB-2516 (2) (IC50 ¼ 3 nM for MMP-9 and IC50 ¼ 6 nM pounds are that these can inhibit other zinc-dependent metal-
for MMP-2) are examples of MMPIs fall under peptidomimetic class loenzymes including ADAMs (a disintegrin and metalloproteinases)
(Fig. 6) [65e69]. such as ADAM-10 and ADAM-17 [58,61]. To address these issues,
Small molecules such as MMI270/CGS-27023A (3) (IC50 ¼ 8 nM next generation MMPIs were developed those includes tanomastat/
for MMP-9 and IC50 ¼ 11 nM for MMP-2), GM6001 (4) (Ki ¼ 0.57 nM BAY12-9566 (7) (IC50 ¼ 301 nM for MMP-9 and IC50 ¼ 11 nM) [76],
for MMP-9 and Ki ¼ 0.39 nM for MMP-2), GI129471 (5) prinomastat/AG-3340 (8) (Ki ¼ 0.26 nM for MMP-9 and
(IC50 ¼ 14.4 nM for MMP-9 and IC50 ¼ 28 nM for MMP-2) and CGS- Ki ¼ 0.05 nM for MMP-2) [18,77] and rebimastat/BMS-275291 (9)
25966 (6) (IC50 ¼ 27 nM for MMP-9 and IC50 ¼ 11 nM for MMP-2) (IC50 ¼ 25 nM for MMP-9 and IC50 ¼ 41 nM for MMP-2) [66] (Fig. 6).
designed as MMPIs in yester years (Fig. 6) [29,65e69]. Among these, tanomastat (7) contains carboxylate group as the
Batimastat (1), the first peptidomimetic MMPI, was found catalytic zinc ion chelator and it is more MMP-2 selective than the
effective for cancer patients (Fig. 6). It exhibited promising effect earlier compounds. It was developed by Bayer Corporation (Pitts-
against several cancer cell lines [70]. It also exhibited significant burgh, PA). It was clinically investigated for the treatment of
anticancer effect as evidenced in numerous preclinical studies and rheumatoid arthritis and solid tumor [29]. In animal studies, it was
number of xenograft and metastatic models [71]. It was better adequately tolerated with mild renal and hepatotoxicity. However,
effective in early stage of cancers as compared to later stage of phase III clinical studies with BAY12-9566 (7) showed inferior
cancers. It also produced synergistic antiproliferative effects in survival time for advanced pancreatic cancer. In addition, it also
combination with docetaxel and captopril [71]. Due to its poor possessed inferior survival against small cell lung cancer patients in
solubility, it was administered through i.v. route. Though it was clinical studies and therefore, a further study with BAY 12e9566 (7)
well-tolerated, it produced mild systemic toxicity along with was postponed [71].
marked abdominal pain and plural infusion [72]. Prinomastat/AG-3340 (8), an optimized version of CGS-27023A
Marimastat (2) was a low-molecular weight non-specific (3), was effective against a variety of MMPs such as MMP-2, -3, -9
S. Mondal et al. / European Journal of Medicinal Chemistry 194 (2020) 112260 9

Fig. 6. Structure of some potent MMP-9 inhibitors (Compound 1e10).

and -13 [77]. It showed effective antiproliferative efficacy against 8. MMP-9 inhibitors
numerous tumor models after oral and intraperitonial dosage
forms. It also resulted in apoptosis and inhibition of tumor growth MMP-9 can be inhibited in different ways either by blocking of
as well as angiogenesis as evidenced in xenograft models of pro-MMP-9 to active MMP-9 conversion or by direct MMP-9 inhi-
different cancers like breast, prostate, colon, non-small cell lung bition or obstruction of the production of the pro-MMP-9 at the
cancer (NSCLS) [66,78]. It also yielded synergistic effect by inhib- transcription level. The macromolecular endogenous inhibitor of
iting tumor growth as well as angiogenesis in combination with MMP-9 is the tissue inhibitor of metalloproteinases (TIMPs) [41].
paclitaxel and carboplatin against NSCLC. It also decreased the TIMPs consist of 184e194 amino acid residues. This can be classified
number of pulmonary metastasis in B16F10 melanoma [66]. How- into two parts N-terminal and C-terminal sub-domains. They are
ever, it was withdrawn from clinical trial because of its lack of ef- divided into four groups (TIMP-1, -2, -3 and -4). Among them,
ficacy and severe toxicities. TIMP-1 bind with the hemopexin domain of pro-MMP-9 to pro-
Rebimastat (9) inhibits MMP-1, -2, -3, -8, -9, -13 and 14. It was duces a complex, thus prevent MMP-9 activation [9]. TIMP-3 has an
studied in breast, prostate, lung, ovarian and pancreatic carcinoma important role in metalloproteinase activities regulation. TIMP acts
[13]. This compound was also withdrawn from clinical trial. by inhibiting the active MMP or by inhibiting the pro-MMP to
Doxycycline hyclate/Periostat® (10), an antibiotic belonging to active MMP conversion. TIMPs are not applied in therapy because
the family of the tetracyclines, is FDA approved drug against peri- of their short half-life. Another endogenous inhibitor is a2 macro-
odontitis [79] (Fig. 6). It is a classic antimicrobial agent of tetracy- globulin [7]. Several proteins including inducing-cysteine-rich
cline derivative studied as anticancer agent in patients. It showed protein with kazal motifs (RECK) suppress MMP-9 [11]. Exact
promising MMP inhibitory activity [5,79]. It also exhibited cytotoxic inhibitory mechanism of these proteins is unknown.
effect against U2OS osteosarcoma, MDA-MB-435 breast and PC-3
prostate cancer cell lines [71].
8.1. Natural MMP-9 inhibitors
Minocycline (11) is a derivative of doxycycline (Fig. 7). Mino-
cycline is a tetracycline derivative which mainly used for the
Some compounds have been identified from natural sources
treatment acne vulgaris and sexually transmitted diseases [41].
mainly flavonoids, alkaloids and phenolic compounds those show
It acts by inhibiting the bacterial 30s ribosomal subunit and thus
MMP inhibition. Polyphenols from Camellia sinensis can inhibit
inhibits protein synthesis. It was found that the non-microbial ac-
MMP-1, -2, -3, -7, -9 thereby prevents photoaging [4]. Ageladine A
tivity of minocycline is due to inhibition of several enzymes
obtained from the marine source sponge Agelas nakamurai can
including MMPs. It inhibits the proteolytic activity of MMP-9 [41].
inhibit MMP-1, -2, -8, -9, -12, 13. Fucoidan extracts from seaweeds
Azithromycin, another compound belongs to the antibiotic class,
Claisiphon novaecaledoniae and methanolic extracts from marine
shows MMP inhibition. It inhibits the gene expression of MMP-9
red algae Cavalina pilulifera show inhibition of MMP-2 and -9
but has no effect on the activation of pro-MMP-9. At high con-
preventing photoaging [4].
centration, azithromycin and minocycline reduces the secretion of
Few natural MMPIs such as silibinin (12) and neovastat (13) are
MMP-9 but have no effect on MMP-2 [41].
reported (Fig. 7) [22,80]. Silibinin, isolated from milk thistle seeds,
10 S. Mondal et al. / European Journal of Medicinal Chemistry 194 (2020) 112260

Fig. 7. Molecular structure of some potent MMP-9 inhibitors (Compound 11e15).

is a flavonoid antioxidant. Being a popular in ethnopharmacognosy 8.2. Monoclonal antibodies as MMP-9 inhibitors
has been used in antihepatotoxic and anti-carcinogenic agents [80].
It is a chemopreventive agent against skin cancer [81]. It has effect Monoclonal antibody (MAbs) has also been developed. MMP-9
on metastasis in breast cancer. It inhibits expression of MMP-9 by targeted MAbs were produced by immunizing mice with recom-
the inhibition of MEK/ERK pathway in a dose dependent manner binant human or mouse MMP-9 proteins [89]. REGA-3G12 is a
[80]. murine monoclonal antibody produced by Hybridoma technology.
An extract from shark cartilage, neovastat/AE-941 (13) (Fig. 7) It acts against MMP-9 by inhibiting the catalytic domain of MMP-9
possesses anti-metastasis and anti-angiogenic effects by inhibiting [90]. It is more specific to MMP-9 because it does not affect MMP-2
vascular endothelial growth factor (VEGF) and MMPs [82]. In [29]. It prevents the mobilization of hematopoietic progenitor cell
phase-I trial, it had shown that neovastat exhibited survival benefit induced by interleukin-8 in rhesus monkeys by inhibitory anti-
at high dose and also well tolerable. The phase-III trial of neovastat bodies against gelatinase B [91].
was started for the patient with colorectal, breast, non-small cell AB0041 is an antibody which inhibits human MMP-9 that found
lung and kidney cancer but the study was terminated later due to to be effective in ulcerative colitis and colorectal cancer [89]. It also
unwanted effects [82]. inhibits rat MMP-9 (rMMP9) but unable to bind to mouse MMP-9
Genistein (14), a soy isoflavonoid, shows anticancer properety (mMMP-9). It is highly MMP-9 selective (more than 500 fold) over
(Fig. 7). It is a potent inhibitor of MMPs through inhibition of tumor other MMP family members. It inhibits the cleavage of gelatine and
growth and invasion [82]. It also possesses estrogenic and anti- thereby prevents the liberation of extracellular matrix sequestered
estrogenic activity. cytokines and growth factors. AB0041 has ability to inhibit the
Gallic acid/GA (15) is a polyhydroxy phenolic compound ob- degradation of MMP-9 mediated epithelial and endothelial base-
tained from various plants, food and fruits (Fig. 7). It has anticancer ment membranes. This antibody also has the ability to inhibit the
activity shown in various cancer cells, leukemia, melanoma, gastric, release of TNF-a thus, expressed anti-inflammatory effects [89].
lung, prostate, breast, cervical and esophageal cancers [83e85]. It Bortezomib is a proteosome inhibitor that inhibits the degra-
acts by decreasing the glutathione level and generating reactive dation of intracellular proteins and causes cell apoptosis. Bortezo-
oxygen species (ROS), thus inducing apoptosis or cancer cell death mib has effect on MMP-2 as well as on MMP-9 at low concentration.
[83]. GA (15) alone and in combination with low-level laser irra- It is used in the treatment of multiple myeloma [41].
diation showed anti-cancer effect on human breast cancer cell line
MDA-MB-231, non-tumorigenic breast epithelial cell line MCF10A, 8.3. Other active MMP-9 inhibitors
and melanoma cell line A375 dermal fibroblast cell line HDF [84]. It
reduces metastasis by downregulating the MMP-2/-9 expression in Elevated expression of MMP-9 is associated with various path-
leukemic cells. However, it becomes toxic to the human micro- ological conditions such as cancer, inflammation, tumor progres-
vascular endothelial cell when treated at a dose of more than sion, atherosclerosis, etc. Because of its crucial role in various
10,000 nM [83]. It has been shown that the hydroxyl group at para- diseases many MMP-9 inhibitor have been developed so far
position to the carboxyl group of GA (15) is important for MMP-2/ [92e118].
MMP-9 downregulating effect [83]. Apart from GA (15), catechins,
i.e., (-)-epigallocatechin-3-gallate (EGCG) and flavonoids (quer- 8.3.1. Hydroxamate based MMP-9 inhibitors
cetin) both have anticancer effect by inhibiting MMP-2/-9 functions Yamamoto et al. [92] reported some hydroxamate-based potent
[83,86,87]. MMPIs. Compound 16 (Fig. 8) was a potent and selective gelatinase
CT1746 is a synthetic MMP-2/-9 inhibitor which is more active inhibitor (MMP-2 IC50 ¼ 1.6 nM, MMP-9 IC50 ¼ 0.2 nM) with a
in combination with cisplatin or cyclophosphamide than the single minimum of 50 fold selectivity over MMP-1 and -3.
dose therapy [88]. It reduces tumour growth and pulmonary Hanessian and co-workers [94] reported some acyclic
metastasis [88]. sulfonamide-based hydroxamates having selective MMP-9
S. Mondal et al. / European Journal of Medicinal Chemistry 194 (2020) 112260 11

inhibitory activities over MMP-1, -3 and -13. However, Compound Fabre et al. [102] reported some triazolyl-substituted arylsul-
17 (Fig. 8) showed potent MMP-2 and -9 inhibition (IC50 ¼ 0.7 nM fonyl hydroxamates as highly selective and potent inhibitors of
for MMP-2 and IC50 ¼ 0.2 nM for MMP-9) with greater selectivity MMP-2 and MMP-9. Compound 23 (Fig. 8) exhibited potent MMP-2
over MMP-3 and -13. (IC50 ¼ 0.50 nM) and MMP-9 inhibition (IC50 ¼ 0.80 nM).
Baxter et al. [96] reported some arylsulfonyl hydroxamates as A series of g-fluorinated a-aminocarboxylic and a-amino-
potent MMPIs. Compound 18 (Fig. 8) among these molecules yiel- hydroxamic acids was developed by Behrends and co-workers
ded potent but non-selective MMP-9 inhibition (IC50 ¼ 4 nM for [103] as potent gelatinase inhibitors. Compound 24 (Fig. 8)
MMP-2 and IC50 ¼ 5 nM for MMP-9). This compound also maintains showed potent MMP-9 inhibition (IC50 ¼ 0.54 nM) having mini-
high degree of selectivity over MMP-1 (IC50 ¼ 2000 nM), MMP-3 mum 7 fold selectivity over MMP-2 (IC50 ¼ 3.9 nM).
(IC50 ¼ 100 nM), MMP-8 (IC50 ¼ 15 nM). Apart from that a signifi- Nuti et al. [104] reported some N-isopropoxy-arylsulfonamido
cant plasma level concentration at 30 mg/kg dose, compound 18 hydroxamates as promising and selective inhibitors of MMP-2 over
displayed 40% inhibition of B16F10 melanoma in mouse model that other MMPs. Though Compound 25 (Fig. 8) showed potent MMP-2
was comparatively similar like marimastat (2). inhibition (IC50 ¼ 0.67 nM), it exhibited comparatively better MMP-
Hanessian et al. [97] reported some arylsulfonyl homocysteine 9 (IC50 ¼ 0.43 nM) and MMP-13 (IC50 ¼ 0.19 nM) inhibitory activ-
hydroxamates as potent MMPIs. Most of these derivatives displayed ities. The in vitro study suggested that compound 25 also signifi-
potent and selective MMP-9 inhibition than other MMPs. However, cantly decreased the invasion and migration of human umbilical
Compound 19 (Fig. 8) yielded potent MMP-2 inhibition vein endothelial cells (HUVEC) at lower concentration. Further-
(IC50 ¼ 0.30 nM) but showed 30 fold better affinity towards MMP-9 more, the western blot and gelatine zymography analysis further
(IC50 ¼ 0.01 nM). proved that Compound 25 had the ability to inhibit gelatinase ac-
Further study conducted by Hanessian and co-workers [98] tivity. It also displayed potential cytotoxicity and apoptosis-
resulted in some arylsulfonamido hydroxamates having MMPs inducing ability in endothelial cells. Moreover, it was also found
inhibitory properties. Compound 20 (Fig. 8) exhibited the highest to respond well to exert antiangiogenic activity in vivo as evidenced
MMP-2 inhibition (IC50 ¼ 1.64 nM) but nearly 2 fold better affinity by the matrigel sponge assay model in mice.
towards MMP-9 (IC50 ¼ 0.9 nM). This compound was non-selective A series of arylsulfonamido hydroxamates was reported as
to MMP-1 (IC50 ¼ 198 nM), MMP-3 (IC50 ¼ 6.7 nM), MMP-13 potent gelatinase inhibitors by Zapico and collegues [105]. Com-
(IC50 ¼ 5.5 nM). pound 26 (Fig. 9) yielded potent MMP-2 inhibition (IC50 ¼ 0.32 nM)
Structural exploration by Yamamoto et al. [99] led to the having 3.4 fold selectivity over MMP-9 (IC50 ¼ 1.06 nM).
development of some N-phenoxy benzyl g-aminobutyric acid Hugenberg et al. [69] reported g-fluorinated sulfonylamino
hydroxamates. Compound 21 (Fig. 8) showed excellent MMP-9 hydroxamic acid derivative based on the lead structure of CGS
inhibition (IC50 ¼ 0.3 nM) having 2.3 fold selectivity over MMP-2 27030A. Compound 27 (Fig. 9) (S-enantiomer) showed potent in-
(IC50 ¼ 0.7 nM). hibition against MMP-9 (IC50 ¼ 3.0 nM) with minimum 10 fold
Yang and co-workers [100] did the development of b-N biar- higher selectivity than MMP-2 (IC50 ¼ 32.8 nM).
ylether sulfonamido hydroxamates as selective and potent MMP-9
inhibitors over MMP-2. Compound 22 (Fig. 8) resulted in potent 8.3.2. Non hydroxamate-based MMP-9 inhibitor
MMP-9 inhibitory activity (IC50 ¼ 0.52 nM) and 25 fold selectivity It has been found that hydroxamate moiety provides toxicity,
over MMP-2 (IC50 ¼ 13 nM). Some a-tetrahydropyranyl-based metabolic instability and also lowers selectivity. For these reasons,
arylsulfonyl hydroxamates were reported as selective and potent non hydroxamate zinc binding groups (carboxylates, N-hydrox-
MMP-2 inhibitors over MMP-9 [101]. yformamides, pyrimidine-2,4,6-triones and others) containing

Fig. 8. Molecular structure of other active MMP-9 inhibitors (Compound 16e25).


12 S. Mondal et al. / European Journal of Medicinal Chemistry 194 (2020) 112260

Fig. 9. Molecular structure of other active MMP-9 inhibitors (Compound 26e35).

compounds were developed [18,106]. potent and selective against MMP-9. It was about 60 fold MMP-9
selective compared to MMP-2 (IC50 ¼ 4.49 nM for MMP-9 and
8.3.2.1. Carboxylic acid based MMP-9 inhibitors. Kiyama et al. [106] IC50 ¼ 266 nM for MMP-2).
developed some arylsulfonamido-based carboxylic acids as selec- Beutel and colleagues [118] showed a number of carboxylic acid
tive and potential MMP-2 inhibitors over MMP-9. Compound 28 and hydroxamic acid based compound exhibited MMP-2 and MMP-
(Fig. 9) showed almost same inhibitory activity towards MMP-2 and 9 inhibitory activity. Among these molecules, compound 35 (Fig. 9)
MMP-9 (IC50 ¼ 4.4 nM for MMP-9 and IC50 ¼ 4.8 nM for MMP-2) having selectivity for both gelatinases (MMP-2 and MMP-9). It has
slightly more active against MMP-9. minimum 12 fold selectivity for MMP-2 than MMP-9 (IC50 ¼ 3.7 nM
Zhang et al. [76] reported a series of biphenylsulfonamido car- for MMP-2 and IC50 ¼ 36.4 nM for MMP-9). It is orally available with
boxylic acids as promising gelatinases inhibitors. However, most of little or no adverse effect.
these compounds of this series yielded selective and potent MMP-2
inhibition compared to MMP-9. Compound 29 (Fig. 9) showed
potent MMP-9 inhibition (IC50 ¼ 0.5 nM) similar with MMP-2 in- 8.3.2.2. N-Hydroxyformamides as MMP-9 inhibitors. Wada and co-
hibition (IC50 ¼ 0.7 nM). workers [112] investigated a series of phenoxyphenyl sulfone N-
Selivanova and co-workers [107] reported some arylsulfona- formylhydroxamates as potent and orally bioavailable MMPIs. S-
mide carboxylic acids having indole function as promising gelati- stereoisomer of compound 36 (Fig. 10) showed promising selective
nases inhibitors. Compound 30 (Fig. 9) displayed potent MMP-2 MMP-9 inhibition (IC50 ¼ 0.092 nM) over MMP-2 (IC50 ¼ 0.36 nM).
and MMP-9 inhibition (IC50 ¼ 0.9 nM for MMP-2 and IC50 ¼ 1 nM
for MMP-9).
Halder et al. [108] reported a series of L-(þ)-isoglutamine de- 8.3.2.3. Pyrimidine-2,4,6-triones as MMP-9 inhibitors. Among
rivatives having dual MMP-2 and histone deacetylase 8 (HDAC8) nitrogen-based heterocyclic ZBGs, pyrimidine-2, 4, 6-trione and
inhibitory activities. Compound 31 (Fig. 9) was the a MMP-2 in- pyrimidine dionethione based inhibitors were mostly studied. As
hibitor (IC50 ¼ 6400 nM) and showed better affinity towards MMP- these inhibitors belonged to the barbiturate class, metabolism and
9 (IC50 ¼ 4830 nM). bioavailability were studied extensively in many FDA-approved
Adhikari and co-workers [109] synthesized and checked the barbiturates [113]. This type of inhibitors coordinates to catalytic
biological activities of some biphenylsulfonyl derivatives. Among Zn2þ ion by the N3 atom of the barbiturate function [114,115]. Some
these compound 32 (Fig. 9) exhibited both MMP-2 and MMP-9 C-5 di-substituted barbiturates were reported as potential gelati-
inhibitory activity but 20.53 times more selective towards MMP-2 nase inhibitors by Breyholz and co-workers [116]. Compound 37
over MMP-9 (IC50 ¼ 24 nM for MMP-2 and IC50 ¼ 492 nM for (Fig. 10) exhibited potential MMP-9 inhibition activity (IC50 ¼ 1 nM)
MMP-9). having 26 fold better activity as well as selectivity against MMP-2
Some pentanoic acid derivatives were reported by the same (IC50 ¼ 26 nM). The 125I-radiolabeled compound 37 was a useful
group showed potent and selective MMP-2 inhibitory activities radio-imaging tool and effective in treating diseases like inflam-
[110]. The compound 33 (Fig. 9) was non-selective but shows mation, atherosclerosis and cancer.
comparatively better affinity towards MMP-9 than MMP-2 Wang et al. [117] reported some N-substituted homopiperazine
(IC50 ¼ 4800 nM for MMP-9 and IC50 ¼ 5340 nM for MMP-2). barbiturates as promising gelatinase inhibitors. Compound 38
Ayoup et al. [111] reported some a-aminoacyl amide derivatives (Fig. 10) yielded similar potency against both MMP-2 and -9
as potent MMP-9 inhibitors. Compound 34 (Fig. 9) was found to (IC50 ¼ 1.1 nM).
S. Mondal et al. / European Journal of Medicinal Chemistry 194 (2020) 112260 13

Fig. 10. Molecular structure of other active MMP-9 inhibitors (Compound 36e38).

9. Trends in the development of potent and selective MMP-9 molecules are either inactive or poorly active against MMP-1 and
inhibitors are non-selective in nature. Though compound 44 (Fig. 11) of this
series was non-selective in MMP-3 and MMP-9 delivered almost
In the year 1998, Levin and his colleagues [119] reported a group 7e2639 fold MMP-9 selectivity over MMP-1, MMP-2 and MMP-13
of hydroxamate containing diazepine derivatives as potent in- [95].
hibitors of MMPs. The in vitro enzyme inhibition assay of these Pikul et al. [123] reported a set of 6-oxo hexahydro pyrimidine
compounds against MMP-1, MMP-9 and MMP-13 not just clearly and [1,4]-diazepane containing hydroxamate derivatives as potent
depicts their potent MMP inhibitory property but also exhibits their MMPIs. The study reveals that half of these pyrimidine derivatives
MMP-9 selective nature. The MMP inhibitory profile of these are inactive against MMP-7 compared to their diazepane analogs.
molecules also shows that the substitutions at the phenyl ring and Although these authors provided a crystal structure study of one of
heterocyclic nitrogen atom play important roles for their potency as those pyrimidine derivatives (compound 45) (Fig. 11) with stro-
well as their MMP-9 selectivity. In this set of MMPIs, compound 39 melysin, the recemic mixture of compound 46 (Fig. 11) delivered
(Fig. 11) showed the highest MMP-9 selectivity over MMP-1 (67.9 more or less 3.6e1810 fold MMP-9 selectivity over MMP-1, MMP-3,
fold) and MMP-13 (2.85 fold) while being highly active against MMP-7 and MMP-8. Compound 46 was also equipotent in MMP-2,
MMP-9 [119]. MMP-9 and rat MMP-13 [123].
A set of potent, selective and orally bioavailable gelatinase in- The discovery of some anthranilic acid derivatives as potent
hibitors were reported by Tamura et al. [120]. The in vitro study of MMPIs were reported by Levin et al. [124]. The in vitro enzyme
these compounds showed them as either non-selective or MMP-2 inhibitory assay of these molecules suggested them as MMP-9/
selective molecules. Compound 40 (Fig. 11) of this series exhibi- MMP-13 non-selective compounds while being selective over
ted almost 4 fold selectivity toward MMP-9 over MMP-2. This MMP-1 and TACE. Compound 47 (Fig. 11), a meta-trifluoromethyl
compound 40 also showed the higher plasma concentration phenyl analog of this series, showed 2-fold, 294 fold and 542 fold
exhibiting a Cmax of 284.5 mM in mice. MMP-9 selectivity over MMP-13, TACE and MMP-1 respectively.
Almsted and colleagues [121] reported a series of thiazine and Additionally, the compound 48 (Fig. 12) with dimethylamino group
thiazepine derivatives as potent MMPIs. Though most of these substitution at the sulfonamido nitrogen atom seemed to produce
molecules exhibited their no-selective nature against the tested the higher MMP-9 selectivity over MMP-13 [124].
MMPs, compound 41 (Fig. 11) of this series delivered 4.8 fold, 2.8 In continuation to the anthranilates as potent MMPIs, Levin et al.
fold, 6.6 fold, 36 fold and 3.8 fold selectivity for MMP-9 over MMP- [125] incorporated basic amines into the structure and reported a
1, MMP-2, MMP-3, MMP-7 and MMP-8 respectively. group of newer and potent anthranilic acid analogs as potent MMP
In order to develop piperazine based MMPIs, Cheng and co- inhibitors. Among these newer anthranilates, compound 49
workers [122] designed and synthesized a pool of hydroxamate (Fig. 12) and compound 50 (Fig. 12) respectively exhibited an
containing piperazine analogs as potent inhibitors of MMPs. approximate range of 3.6e81 fold and 2.8e121.6 fold MMP-9
Although most of these compounds showed their non-selective selectivity over MMP-1, MMP-13 and TACE [125].
nature between MMP-9 and MMP-13, these MMPIs expressed Nelson and co-workers [126] developed a set of potent benzo-
their higher selectivity toward MMP-9 compared to MMP-1, MMP- diazepine analogs as potent MMP and TACE inhibitors and tested
3 and MMP-7. Compound 42 (Fig. 11) of this series displayed potent those compounds against MMP-1, MMP-9, MMP-13 and TACE
MMP-9 inhibition in picomolar concentration while being almost in vitro. Although most of these inhibitors showed higher inhibitory
9e805 fold MMP-9 selective over MMP-1, MMP-3 and MMP-7. potency against all these tested metalloenzymes, compound 51
Compound 42 was also non-selective to MMP-13. Additionally, (Fig. 12) containing para-methoxy phenyl sulfonyl and N-methyl
compound 43 (Fig. 11), N-carboxy pyridine analogue of compound carboxymethyl piperazine groups at the diazepine nitrogen atoms
42, was almost 2 fold selective to MMP-9 over MMP-13 and 6e865 exhibited enzyme inhibition along with higher MMP-9 selectivity
fold selective over MMP-1, MMP-3 and MMP-7 [122]. (1.69e26.64 fold) over MMP-1, MMP-13 and TACE [126].
With intentions to develop specific MMPIs, Chollet et al. [95] A series of retro hydroxamate derivatives were reported by
synthesized a group of 3-bis-aryloxy propionic acid derivatives as Wada et al. [112] as potent, selective and orally bioavailable in-
potent MMPIs. The in vitro studies suggest that most of these hibitors of MMPs. These sulfone-N-formyl hydroxamates seemed to
14 S. Mondal et al. / European Journal of Medicinal Chemistry 194 (2020) 112260

Fig. 11. Molecular structure of MMP-9 inhibitors (Compound 39e47).

Fig. 12. Molecular structure of MMP-9 inhibitors (Compound 48e56).

be non-selective against MMP-2 and MMP-9 but most of these Zask et al. [127] synthesized and reported some phenyl sulfo-
molecules showed inactivity against MMP-1. Compound 52 (Fig. 12) namido and hydroxamate containing quinolone and hetero biaryl
of this series showed greater than 2 fold MMP-9 selectivity over derivatives as potent MMP and TACE inhibitors. The in vitro studies
MMP-2 and 241 fold selectivity over MMP-1 while being able to showed that most of these molecules were highly potent against
show a good bioavailability in cynomologus monkey [112]. MMP-9 and MMP-13 but were comparatively less potent against
S. Mondal et al. / European Journal of Medicinal Chemistry 194 (2020) 112260 15

MMP-1 and TACE. Although bearing a MMP-9/MMP-13 non-se- inhibition. This Compound 60 also exhibited a range of 2 to
lective nature, compound 53 (Fig. 12) exhibited 2e387.5 fold MMP- 45,000 fold MMP-9 selectivity over MMP-1, MMP-2, MMP-3, MMP-
9 selective nature over MMP-1, MMP-13 and TACE [127]. 8, MMP-13 and MMP-14 [133].
Some novel arylsulfone piperidine-4-hydroxamate derivatives A few novel Cobactin-T based molecules were reported by
were reported by Aranapakam et al. [128] as potent and orally active Wilson et al. [134] as potent MMPIs. The in vitro study reveals that
MMPIs for the treatment of osteoarthritis. The in vitro enzyme most of these molecules are highly potent and dual MMP-2/MMP-9
inhibitory activity studies of these molecules clearly depict their dual active in nature. Compound 61 (Fig. 13) of this series showed almost
MMP-9/MMP-13 selective nature over MMP-1 and TACE. Though 2 fold, 409 fold and 3.1 fold MMP-9 selectivity than MMP-2, MMP-3
these MMPIs showed comparative higher affinity for MMP-13 over and MMP-13, respectively [134].
MMP-9, compound 54 (Fig. 12) with para-n-butyloxy phenyl sulfonyl Through solid phase and in silico evaluation approaches Topai
and N-benzyl substitutions was approximately 5 fold and 92 fold et al. [135] synthesized a group of potent inhibitors of MMP-2 and
selective to MMP-9 than MMP-13 and TACE respectively. Compound MMP-9. A large number of these compounds either expressed their
54 was also poorly active against MMP-1 and expressing 48% MMP-1 selectivity toward MMP-2 over MMP-9 or were non-selective in
inhibition at 10 mM concentrations in vitro [128]. MMP-2 and MMP-9. In this scenario, compound 62 (Fig. 13) of this
In a stereo specific synthetic approach, Diguarher et al. [129] series exhibited 4.5 fold MMP-9 selectivity over MMP-2.
developed a set of bis-aryl cyclopentane based carboxylic acid de- A group of potent triazole-based hydroxamate derivatives were
rivatives as specific and potent MMP inhibitors. The in vitro study synthesized and biologically evaluated by Hugenberg et al. [136].
suggests these compounds as poorly active MMP-1 inhibitors and The in vitro MMP inhibitory assay of these molecules significantly
reveals their MMP-2 selective nature. Compound 55 (Fig. 12) of this indicates that the ClogD values of these molecules might have a
series showed promising MMP-9 selectivity of 3.5e3846 fold over correlation with their MMP inhibitory potency. Moreover, com-
MMP-1, MMP-2, MMP-3 and MMP-13 [129]. pound 63 (Fig. 13) of this series displayed a range of 2.5 folde7 fold
In search of the treatment of ischemic stroke Zhang et al. [130] MMP-9 selectivity over the other tested MMP isoforms in vitro
developed some pyridinone derivatives as potent MMP inhibitors. [136].
Most of these compounds showed dual MMP-2/MMP-9 selective in With the intentions to develop selective MMP-2 inhibitors,
nature at sub-nanomolar range in vitro. A few molecules of this Kreituss and co-workers [137] reported a set of aziridine-triazole
series were also inactive in MMP-1 and produced an activity range conjugates as potent and selective MMP-2 inhibitors. Regardless
of 0.87e79.1 nM in MMP-2, MMP-3, MMP-9 and MMP-13. From of their MMP-2 selective nature, compound 64 (Fig. 13) showed
these few molecules, compound 56 (Fig. 12) delivered 6.43 fold, almost 30% inhibition of MMP-9 at 20 mM concentrations and was
7.82 fold and almost 91 fold MMP-9 selectivity over MMP-2, MMP- also better active in MMP-9 over MMP-3, MMP-7, MMP-12, MMP-
13 and MMP-3 respectively [130]. 13 and MMP-14 in vitro.
Through optimization of substituents, Yang and co-workers In a study to develop promising MMPIs as anticancer agents for
[100] reported a group of biaryl ether sulfonamido hydroxamate angiogenesis, Hu et al. [138] synthesized a few peptides as potent
derivatives as potent gelatinase inhibitors. The MMP inhibitory anticancer agents. The in vitro study of these peptides depicts that
activities of these molecules suggest that the special configuration the one of these peptides [CPU-1: Pro-3-(3-pyridyl)-L-alanine-Cys-
of these molecules may have a correlation with their MMP-9 L-4,40 -biphenylalanine-Arg-Gly-Glu-Gly-Gly-Gly-Gly-Ile-Val-Arg-
selectivity. The compound 57 (Fig. 13) of this series expressed the Arg-Ala-Asp-Arg-Ala-Ala-Val-Pro] showed 1.5 fold and 19 fold
highest MMP-9 selectivity over MMP-2 when compared to other selectivity toward MMP-9 over MMP-3 and MMP-8 respectively.
molecules of this series along with a picomolar range MMP-9 in- These authors also reported that the polypeptide “CPU-1” was able
hibition in vitro [100]. to significantly inhibit the growth of B16F10 mouse melanoma cell
A group of sulfonamide derivatives as broad-spectrum MMPIs line [138].
were reported by Wagner and colleagues [131] as a tool of molec- Some potent g-fluorinated amino hydroxamates and amino
ular imaging. Beside their non-selective nature, these molecules carboxylic acid derivatives were reported by Behrends et al. [103].
expressed higher affinity toward either MMP-2 or MMP-13. Com- The in vitro study of these molecules transparently exhibited the
pound 58 (Fig. 13) of this series, containing a carboxylic acid group superiority of hydroxamic acid derivatives over the carboxylic acid
and a para-fluoro-ethoxy phenyl sulfone moiety showed promising analogs for higher MMP inhibition. Interestingly, the chirality/
MMP-9 selectivity over MMP-1, MMP-2 and MMP-13. Although special-conformation of these compounds may have a significant
compound 58 is 26 fold less potent against MMP-9 when compared amount of influence on their activity. In most of these cases for the
to its hydroxamate analog, it was almost 2 fold and 4.3 fold MMP-9 carboxylic acid derivatives, the (R)-configured compounds deliv-
selectivity over MMP-2 and MMP-1 but was also inactive in MMP- ered poor MMP inhibition (for both MMP-2 and MMP-9) whereas
13 [131]. the (S)-enantiomers of those molecules were unable to deliver any
A series of selective and potent MMP-12 inhibitors were MMP inhibition. The compound 65 (Fig. 13) ((S)-2) of this series
designed and synthesized by Nuti et al. [132]. The in vitro biological showed almost 11 fold more MMP-9 selectivity over MMP-2. The
evaluation of these molecules against a broad spectrum of MMPs (R)-conformer of compound 65 was 4 fold less potency in MMP-9
clearly indicates the benzoic hydroxamates of this series as non- and 2 fold more MMP-2 selectivity when compared to compound
selective MMPIs. On the other hand, the phenyl acetic hydrox- 65 (Fig. 13 [103].
amate analogs of these series were both potent and selective to JNJ0966 (66) is inhibited the activation of MMP-9 zymogen [7]
MMP-12. Compound 59 (Fig. 13), a 4-(para-methoxyphenyl)-oxy- (Fig. 13). It inhibits the activation of MMP-9 by preventing the
phenyl sulfonyl group containing benzoic hydroxamate derivative conversion of pro-MMP-9 into catalytically active MMP-9 [7].
showed an approximate range of 2e54 fold MMP-9 selectivity over Scannevin et al. [7] reported the effect of JNJ0966 to inhibit MMP-9
MMP-2, MMP-3, MMP-8, MMP-12, MMP-13, MMP-14 and MMP-16 activation.
[132].
Some hydroxamate group containing a-tetrahydropyranyl sul- 10. Conclusion and future perspective
fone and a-piperidine sulfone derivatives as MMPIs were reported
by Becker et al. [133]. The N-2-pyridyl methyl piperidine containing Physiological and pathophysiological functions of body depend
compound 60 (Fig. 13) showed potent and selective MMP-9 on the impact of proteolytic enzymes function. Therefore, broad
16 S. Mondal et al. / European Journal of Medicinal Chemistry 194 (2020) 112260

Fig. 13. Molecular structure of MMP-9 inhibitors (Compound 57e66).

spectrum inhibition or blockade of protease function will reveal allows stronger van der Waals interaction compared to the cor-
some systemic effects. These effects may be beneficial, neutral and responding S2 subsite of MMP-2. There are some specific van der
harmful to the body due to inhibition of tumor cell invasion and Waals interactions with the indole moiety also (interactions with
metastasis. Therefore, research on new agent with anticancer amino acid residues Phe421 and Met422). Again, incorporation of
property is regained. MMP-9 has important role in ECM remodel- phenyl ring at the 2nd position of the indole moiety of the ligand
ling, metastasis, angiogenesis, apoptosis and cancer progression. structure displayed some specific van der Waals interaction with
Hence, MMP-9 and its inhibitors could be important target for Leu187 and His411 at the S2’ subsite of MMP-9. These specific
anticancer drug. Selective inhibition of these enzymes in particular interactions have been found to be important during designing
site is linked with potential anticancer activity. Depending on the selective MMP-9 inhibitors [139]. Khandelwal and colleagues
structural studies many MMP-9 inhibitor have been to be designed [140] performed a combined quantum mechanics/molecular
used in the treatment of cancer. It has been found that most of the mechanics (QM/MM), molecular dynamics (MD) simulation and
MMP-9 inhibitors were not selective and showed little efficacy. 3D-QSAR study on some MMP-9 inhibitors. These approaches also
These can also inhibit other MMPs with similar affinity may be effective in designing selective MMP-9 inhibitors. Never-
simultaneously. theless, receptor-based 3D-QSAR study performed by Tuccinardi
The structural resemblance of MMP-9 with other MMP iso- et al. [141] identified the role of some hydrophobic amino acid
forms makes it quite difficult (especially medium size S1’ pocket residues namely Tyr237 and Tyr420 those are responsible for
MMPs namely MMP-2, MMP-8, MMP-9 and MMP-12). However, stabilizing the substrate at the binding site. QSAR study per-
MMP-9 inhibitors may be highly useful in anticancer therapeutics formed by Ramezani and Shamsara [142] revealed that selective
if selective MMP-9 inhibitors are designed. This will also allow MMP-9 inhibitors may be designed. Gao et al. [143] performed a
restricting the off-target adverse effects. Among various ap- ligand-based pharmacophore mapping and molecular docking
proaches, molecular modelling aspects may be considered to gain analysis to screen selective MMP-9 inhibitors. Jana and Singh
a greater acceptance for reducing time and money. Amino acid [144] identified some selective MMP-9 inhibitors through the
residues forming the S1’ pocket play a major role regarding combined study of pharmacophore mapping and 3D-QSAR fol-
selectivity issue is concerned. Nevertheless, the type of hydro- lowed by virtual screening and drug-likeliness filtering on ADME
phobic aryl group present in the ligand/drug structure directed to properties. Some selective MMP-9 inhibitors were screened from
fit the S1’ pocket is also equally important for the selective inhi- natural products by Hou and co-workers [145] through ligand-
bition. Therefore, both the ligand structure and the amino acid based pharmacophore mapping followed by molecular docking,
residues forming different pockets (namely, S1’, S1 and S2) may be MD simulation as well as bioassay and assessment of ADMET
identified by molecular docking study. Molecular docking study studies. Apart from these molecular modelling studies, other
performed by Ledour et al. [139] showed that van der Waals methodologies such as quantitative activity-activity relationship
interaction between the p-bromobiphenyl group and Phe110 at S2 (QAAR) may also be utilized to design selective MMP-9 inhibitors
subsite is crucial for MMP-9 selectivity compared to the van der [146-147]. Depending on these molecular modelling techniques,
Waals interaction of the same structural motif with Asn111 of newer active as well as selective MMP-9 inhibitors targeted to
MMP-2. According to that study, the smaller S2 subsite of MMP-9 anticancer therapy may be designed.
S. Mondal et al. / European Journal of Medicinal Chemistry 194 (2020) 112260 17

Declaration of interest statement [16] A. Yabluchanskiy, M. Yonggang, R.P. Iyer, M.E. Hall, M.L. Lindsey, Matrix
metalloproteinase-9: many shades of function in cardiovascular disease, Am.
Physiol. Soc. 28 (2013) 391e403.
The authors have no conflict of interests. [17] O. Nyormoi, L. Mills, M. Bar-Eli, An MMP-2/MMP-9 inhibitor, 5a, enhances
apoptosis induced by ligand of the TNF receptor super family in cancer cells,
Cell Death Differ. 10 (2003) 558e569.
Acknowledgement [18] J.A. Jacobsen, J.L.M. Jourden, M.T. Miller, S.M. Cohen, To bind zinc or not to
bind zinc: an examination of innovative approaches to improved metal-
loproteinase inhibition, Biochim. Byophys. Acta. 1803 (2010) 72e94.
SM is thankful to the All India Council for Technical Education
[19] N. Adhikari, S.A. Amin, T. Jha, Collagenases and gelatinases and their in-
(AICTE) New Delhi for awarding a fellowship. NA is grateful to hibitors as anticancer agents, in: S.P. Gupta (Ed.), Cancer Leading Proteases,
Council of Scientific and Industrial Research, India (CSIR), New Elsevier, 2020, pp. 265e294.
[20] M. Fanjul-Fern  pez-Otín, Matrix met-
andez, A.R. Folgueras, S. Cabrera, C. Lo
Delhi, India for providing research associateship (RA) [FILE NO.: 09/
alloproteinases: evolution, gene regulation and functional analysis in mouse
096(0966)/2019-EMR-I, Dated: 28-03-2019]. SB is grateful to the models, Biochim. Biophys. Acta 1803 (2010) 3e19.
RUSA 2.0 of University Grant Commission (UGC), New Delhi, India [21] R.T. Aimes, J.P. Quigley, Matrix metalloproteinase-2 is an interstitial colla-
to Jadavpur University, Kolkata, India for awarding fellowship [REF. genase: inhibitor-free enzyme catalyzes the cleavage of collagen fibrils and
soluble native type I collagen generating the specific 3/4- and 1/4-length
NO: R-11/1030/19, Dated: 30-07-2019]. SAA sincerely acknowl- fragments, J. Biol. Chem. 270 (1995) 5872e5876.
edges CSIR, New Delhi, India for awarding the Senior Research [22] I. Gkouveris, N.G. Nikitakis, J. Aseervatham, N. Rao, K.U.E. Ogbureke, Matrix
Fellowship (SRF) [FILE NO.: 09/096(0967)/2019-EMR-I, Dated: 01- metalloproteinases in head and neck cancer: current perspectives, Metal-
loproteinases Med. 4 (2017) 47e61.
04-2019]. TJ is thankful for the financial support from RUSA 2.0 of [23] C. Tallant, A. Marrero, F. Gomis-Rüth, Matrix metalloproteinases: fold and
UGC New Delhi to Jadavpur University, Kolkata, India. The authors function of their catalytic domains, Biochim. Biophys. Acta 1803 (2010)
heartily thank Dr. Balaram Ghosh of BITS Pilani, Hyderabad 20e28.
[24] Y. Itoh, Membrane-type matrix metalloproteinases: their functions and
Campus, India as well as Mr. Sandip Kr. Baidya, Mr. Saptarshi Sanyal regulations, Matrix Biol. 46 (2015) 207e223.
and Mr. Rajat Sarkar of Jadavpur University, Kolkata for their kind [25] P.E. Van den Steen, B. Dubois, I. Nelissen, P.M. Rudd, R.A. Dwek,
support. The authors also thank the support from Department of G. Opdenakker, Biochemistry and molecular biology of gelatinase B or matrix
metalloproteinase-9 (MMP-9), Crit. Rev. Biochem. Mol. Biol. 37 (2002)
Pharmaceutical Technology, Jadavpur University, Kolkata, India for
375e536.
providing the research facilities. [26] N. Sela-Passwell, G. Rosenblum, T. Shoham, I. Sagi, Structural and functional
bases for allosteric control of MMP activities: can it pave the path for se-
lective inhibition? Biochim. Biophys. Acta 1803 (2010) 29e38.
References [27] H.E. Van Wart, H. Birkedal-Hansen, The cysteine switch: a principle of
regulation of metalloproteinase activity with potential applicability to the
[1] Cancer- World Health Organization, 2018. (Accessed 12 September 2019). entire matrix metalloproteinase gene family, Proc. Natl. Acad. Sci. U. S. A 87
[2] H.T. Vu, T.X. Hoang, J.Y. Kim, All-trans retinoic acid enhances matrix metal- (1990) 5578e5582.
loproteinase 2 expression and secretion in human myeloid leukemia THP-1 [28] E. Morgunova, A. Tuuttila, U. Bergmann, M. Isupov, Y. Lindqvist, G. Schneider,
cells, BioMed Res. Int. (2018), https://doi.org/10.1155/2018/5971080. K. Tryggvason, Structure of human pro-matrix metalloproteinase-2: activa-
[3] T. Djuric, M. Zivkovic, Overview of MMP Biology and Gene Associations in tion mechanism revealed, Science 284 (1999) 1667e1670.
Human Diseases, the Role of Matrix Metalloproteinase in Human Body Pa- [29] J. Cathcart, A. Pulkoski-Gross, J. Cao, Targeting matrix metalloproteinases in
thologies, Francesco Travascio, IntechOpen, 2017, https://doi.org/10.5772/ cancer: bringing new life to old disease, Genes Dis 2 (2015) 26e34.
intechopen.70265. Available from: https://www.intechopen.com/books/the- [30] G. Klein, E. Velleng, M.W. Fraaije, W.A. Kampsa, E.S.J.M. de Bont, The possible
role-of-matrix-metalloproteinase-in-human-body-pathologies/overview-of- role of matrix metalloproteinase (MMP)-2 and MMP-9 in cancer, e.g. acute
mmp-biology-and-gene-associations-in-human-diseases. leukemia, Crit. Rev. Oncol. Hematol. 50 (2004) 87e100.
[4] P. Gupta, Natural products as inhibitors of matrix metalloproteinases, Nat. [31] H. Nagase, F. Woessner, Matrix metalloproteinases, J. Biol. Chem. 274 (1999)
Prod. Chem. Res. 4 (2016) 1. 21491e21494.
[5] A. Jablonska-Trypuc, M. Matejczyk, S. Rosochacki, Matrix metalloproteinases [32] N. Adhikari, A. Mukherjee, A. Saha, T. Jha, Arylsulfonamide and selectivity of
(MMPs), the main extracellular matrix (ECM) enzymes in collagen degra- matrix metalloproteinase-2: an overview, Eur. J. Med. Chem. 129 (2017)
dation, as a target for anticancer drugs, J. Enzym. Inhib. Med. Chem. 31 72e109.
(2016) 177e183. [33] A. Gimeno, R. Beltra n-Debo n, M. Mulero, G. Pujadas, S. Garcia-Vallve, Un-
[6] C. Gialeli, A.D. Theocharis, N.K. Karamanos, Roles of matrix metal- derstanding the variability of the S1’ pocket to improve matrix metal-
loproteinases in cancer progression and their pharmacological targeting, loproteinase inhibitor selectivity profiles, Drug Discov. Today 25 (2020)
FEBS J. 278 (2011) 16e27. 38e57.
[7] R.H. Scannevin, R. Alexander, T.M. Haarlander, S.L. Burke, M. Singer, C. Huo, [34] B.G. Rao, Recent developments in the design of specific matrix metal-
Y. Zhang, D. Maguire, J. Spurlino, I. Deckman, K.I. Carroll, F. Lewandowski, loproteinase inhibitors aided by structural and computational studies, Curr.
E. Devine, K. Dzordzorme, B. Tounge, C. Milligan, S. Bayoumy, R. Williams, Pharmaceut. Des. 11 (2005) 295e322.
C. Schalk-Hihi, K. Leonard, P. Jackson, M. Todd, L.C. Kuo, K.J. Rhodes, Dis- [35] C. Zhang, S.K. Kim, Matrix metalloproteinase inhibitors (MMPIs) from marine
covery of a highly selective chemical inhibitor of matrix metalloproteinase-9 natural products: the current situation and future prospects, Mar. Drugs 7
(MMP-9) that allosterically inhibits zymogen activation, J. Biol. Chem. 292 (2009) 71e84.
(2017) 17963e17974. [36] J.E. Rundhaug, Matrix metalloproteinases, angiogenesis, and cancer, Clin.
[8] H. Nagase, R. Visse, G. Murphy, Structure and function of matrix metal- Canc. Res. 9 (2003) 551e554.
loproteinases and TIMPs, Cardiovasc. Res. 69 (2006) 562e573. [37] C. Tallant, A. Marrero, F. Xavier Gomis-Ruth, Matrix metalloproteinases: fold
[9] H. Huang, Matrix metalloproteinase-9 (MMP-9) as a cancer biomarker and and function of their catalytic domains, Biochim. Biophys. Acta 1803 (2010)
MMP-9 biosensors: recent advances, Sensors 18 (2018) 3249. 20e28.
[10] S.B. Thorsen, S.L. Christensen, S.O. Würtz, M. Lundberg, B.S. Nielsen, [38] W. Wang, C.J. Schulze, W.L. Suarez-Pinzon, J.R. Dyck ., G. Sawicki, R. Schulz,
L. Vinther, M. Knowles, N. Gee, S. Fredriksson, S. Møller, N. Brünner, Intracellular action of matrix metalloproteinase-2 accounts for acute
A.S. Schrohl, J. Stenvang, Plasma levels of the MMP-9:TIMP-1 complex as myocardial ischemia and reperfusion injury, Circulation 106 (2002)
prognostic biomarker in breast cancer: a retrospective study, BMC Canc. 13 1543e1549.
(2013) 598. [39] M. Toth, I. Chvyrkova, M.M. Bernardo, S. Hernandez-Barrantes, R. Fridman,
[11] S.A. Amin, N. Adhikari, T. Jha, Is dual inhibition of metalloenzymes HDAC-8 Pro-MMP-9 activation by the MT1-MMP/MMP-2 axis and MMP-3: role of
and MMP-2 a potential pharmacological target to combat haematological TIMP-2 and plasma membranes, Biochem. Biophys. Res. Commun. 308
malignancies? Pharmacol. Res. 122 (2017) 8e19. (2003) 386e395.
[12] G.B. Fields, New strategies for targeting matrix metalloproteinases, Matrix [40] N. Ramos-DeSimone, E. Hahn-Dantona, J. Sipley, H. Nagase, D.L. French,
Biol. 44e46 (2015) 239e246. J.P. Quigle, Activation of matrix metalloproteinase-9 (MMP-9) via a
[13] A. Winer, S. Adams, P. Mignatti, Matrix metalloproteinase inhibitors in converging plasmin/stromelysin-1 cascade enhances tumor cell invasion,
cancer therapy: turning past failures into future successes, Mol. Canc. J. Biol. Chem. 274 (1999) 13066e13076.
Therapeut. 17 (2018) 1147e1155. [41] J. Vandooren, S. Knoops, Joao L. Aldinucci Buzzo, L. Boon, E. Martens,
[14] E. Bronisz, I. Kurkowska-Jastrzebska, Matrix metalloproteinase 9 in epilepsy: G. Opdenakker, E. Kolaczkowska, Differential inhibition of activity, activation
the role of neuroinflammation in seizure development, Mediat. Inflamm. 2 and gene expression of MMP-9 in THP-1 cells by azithromycin and mino-
(2016) 1e14. cycline versus bortezomib: a comparative study, PloS One 12 (2017) 1e19.
[15] Y. Liu, H. Liu, X. Luo, J. Deng, Y. Pan, H. Liang, Overexpression of SMYD3 and [42] J. Christensen, V.P. Shastri, Matrix-metalloproteinase-9 is cleaved and acti-
matrix metalloproteinase-9 are associate with poor prognosis of patients vated by Cathepsin K, BMC Res. Notes 8 (2015) 1284e1288.
with gastric cancer, Tumor Biol. 36 (2015) 4377e4386. [43] M. Egeblad, Z. Werb, New functions for the matrix metalloproteinases in
18 S. Mondal et al. / European Journal of Medicinal Chemistry 194 (2020) 112260

cancer progression, Nat. Rev. Canc. 2 (2002) 161e174. [69] V. Hugenberg, M. Behrends, S. Wagner, S. Hermann, M. Schafers, H.C. Kolb,
[44] A. Noel, M. Jost, E. Maquoi, Matrix metalloproteinases at cancer tumor-host K. Szardenings, C. Joseph, W. Luis, F. Gomez, K. Kopka, G. Haufe, Synthesis,
interface, Semin. Cell Dev. Biol. 19 (2008) 52e60. radiosynthesis, in vitro and first in vivo evaluation of a new matrix metal-
[45] V.V. Dragutinovic, S.N. Radovanovic, T.L. Izrael-Zivkovic, M.M. Vrvic, Detec- loproteinase inhibitor based on g-fluorinated a-sulfonylaminohydroxamic
tion of gelatinase B activity in serum of gastric cancer patients, World J. acid, EJNMMI Radiopharm. Chem 3 (2018) 1.
Gastroenterol. 12 (2006) 105e109. [70] P.D. Brown, Clinical studies with matrix metalloproteinase inhibitors,
[46] P. Chiranjeevi, K.M. Spurthi, N.S. Rani, G.R. Kumar, T.M. Aiyengar, J.Pathol.Microb. Immunol. 107 (1999) 174e180.
M. Saraswati, G. Srilatha, G.K. Kumar, S. Sinha, C.S. Kumari, B.N. Reddy, [71] M. Hidalgo, S.G. Eckhardt, Development of matrix metalloproteinase in-
S. Vishnupriya, H.S. Rani, Gelatinase B (-1562C/T) polymorphism in tumor hibitors in cancer therapy, J. Natl. Cancer Inst. 93 (2001) 178e193.
progression and invasion of breast cancer, Tumor Biol. 35 (2014) 1351e1356. [72] V.M. Macaulay, K.J. O’Byrne, M.P. Saunders, J.P. Braybrooke, L. Long,
[47] L.J. van’t Veer, H. Dai, M.J. van de Vijver, Y.D. He, A.A. Hart, M. Mao, F. Gleeson, Phase I study of intrapleural batimastat (BB-94), a matrix met-
H.L. Peterse, K. van der Kooy, M.J. Marton, A.T. Witteveen, G.J. Schreiber, alloproteinase inhibitor, in the treatment of malignant pleural effusions, Clin.
R.M. Kerkhoven, C. Roberts, P.S. Linsley, R. Bernards, S.H. Friend, Gene Canc. Res. 5 (1999) 513e520.
expression profiling predicts clinical outcome of breast cancer, Nature 415 [73] S.M. Wojtowicz-Praga, R.B. Dickson, M.J. Hawkins, Matrix metalloproteinase
(2002) 530e536. inhibitors, Invest. N. Drugs 15 (1997) 61e75.
[48] J. Niu, X. Gu, J. Turton, C. Meldrum, E.W. Howard, M. Agrez, Integrin-medi- [74] A.R. Nelson, B. Fingleton, M.L. Rothenberg, L.M. Matrisian, Matrix metal-
ated signaling of gelatinase B secretion in colon cancer cells, Biochem. Bio- loproteinase: biologic activity and clinical implications, J. Clin. Oncol. 18
phys. Res. Commun. 249 (1998) 287e291. (2000) 1135e1149.
[49] M. Agrez, X. Gu, J. Turton, C. Meldrum, J. Niu, T. Antalis, E.W. Howard, The [75] C.M. Overall, C. Lopez-Otin, Strategies for MMP inhibition in cancer: in-
alpha v beta 6 integrin induces gelatinase B secretion in colon cancer cells, novations for the post-trial era, Nat. Rev. Canc. 2 (2002) 657e672.
Int. J. Canc. 81 (1999) 90e97. [76] Y.-M. Zhang, X. Fan, B. Xiang, D. Chakravarty, R. Scannevin, S. Burke,
[50] M. Bjorklund, E. Koivunen, Gelatinase-mediated migration and invasion of P. Karnachi, K. Rhodes, P. Jackson, Synthesis and SAR of alpha-
cancer cells, Biochim. Biophys. Acta 1755 (2006) 37e69. sulfonylcarboxylic acids as potent matrix metalloproteinase inhibitors, Bio-
[51] S. Hiratsuka, K. Nakamura, S. Iwai, M. Murakami, T. Itoh, H. Kijima, org. Med. Chem. Lett 16 (2006) 3096e3100.
J.M. Shipley, R.M. Senior, M. Shibuya, MMP9 induction by vascular endo- [77] D.R. Shalinsky, J. Brekken, H. Zou, C.D. McDermott, P. Forsyth, D. Edwards,
thelial growth factor receptor-1 is involved in lung specific metastasis, Canc. S. Margosiak, S. Bender, G. Truitt, A. Wood, N.M. Varki, K. Appelt, Broad
Cell 2 (2002) 289e300. antitumor and antiangiogenic activities of AG3340, a potent and selective
[52] T. Itoh, M. Tanioka, H. Matsuda, H. Nishimoto, T. Yoshioka, R. Suzuki, MMP inhibitor undergoing advanced oncology clinical trials, Ann. N. Y. Acad.
M. Uehira, Experimental metastasis is suppressed in MMP-9-deficient mice, Sci. 878 (1999) 236e270.
Clin. Exp. Metastasis 17 (1999) 177e181. [78] O. Santos, C.D. McDermott, R.G. Daniels, K. Appelt, Rodent pharmacokinetic
[53] J. Paupert, V. Mansat-De Mas, C. Demur, B. Salles, C. Muller, Cell-surface and anti-tumor efficacy studies with a series of synthetic inhibitors of matrix
MMP-9 regulates the invasive capacity of leukemia blast cells with mono- metalloproteinases, Clin. Exp. Metastasis 15 (1997) 499e508.
cytic features, Cell Cycle 7 (2008) 1047e1053. [79] E.P. Samartzis, D. Fink, M. Stucki, P. Imesch, Doxycycline reduces MMP-2
[54] S. Monferran, J. Paupert, S. Dauvillier, B. Salles, C. Muller, The membrane activity and inhibits invasion of 12Z epithelial endometriotic cells as well
form of the DNA repair protein Ku interacts at the cell surface with metal- as MMP-2 and -9 activity in primary endometriotic stromal cells in vitro,
loproteinase 9, EMBO J. 23 (2004) 3758e3768. Reprod. Biol. Endocrinol. 17 (2019) 38.
[55] J.K. Rybakowski, Matrix metalloproteinase-9 (MMP9)-A mediating enzyme [80] S. Kim, S.H. Kim, S.M. Hur, S.K. Lee, W.W. Kim, J.S. Kim, J.H. Kim, J.H. Choe,
in cardiovascular disease, cancer, and neuropsychiatric disorders, Cardiovasc. S.J. Nam, J.E. Lee, J.H. Yang, Silibinin prevents TPA-induced MMP-9 expres-
Psychiat. Neurol. 2009 (2009) 904836. sion by down-regulation of COX-2 in human breast cancer cells,
[56] A. Tokito, M. Jougasaki, Matrix metalloproteinases in non-neoplastic disor- J. Ethnopharmacol. 126 (2009) 252e257.
ders, Int. J. Mol. Sci. 17 (2016), https://doi.org/10.3390/ijms17071178. [81] R.P. Singh, R. Agarwal, Flavonoid antioxidant silymarin and skin cancer,
[57] Z. Gu, J. Cui, S. Brown, R. Fridman, S. Mobashery, A.Y. Strongin, S.A. Lipton, Antioxidants Redox Signal. 4 (2002) 655e663.
A highly specific inhibitor of matrixmetalloproteinase-9 rescues laminin [82] W. Li, S. Saji, F. Sato, M. Noda, M. Toil, Potential clinical applications of matrix
from proteolysis and neurons from apoptosis in transient focal cerebral metalloproteinase inhibitors and their future prospects, Int. J. Biol. Markers
ischemia, J. Neurosci. 25 (2005) 6401e6408. 28 (2013) 117e130.
[58] V. Mohan, D. Talmi-Frank, V. Arkadash, N. Papo, I. Sagi, Matrix metal- [83] Y.J. Chen, L.S. Chang, Gallic acid down regulates matrix metalloproteinase-2
loproteinase protein inhibitors: highlighting a new beginning for metal- (MMP-2) and MMP-9 in human leukemia cells with expressed Bcr/Abl, Mol.
loproteinases in medicine, Metalloproteinases Med. 3 (2016) 31e47. Nutr. Food Res. 56 (2012) 1398e1412.
[59] A. Churg, R. Wang, X. Wang, P.O. Onnervik, K. Thim, J.L. Wright, Effect of an [84] K. Khorsandi, Z. Kianmehr, Z. Hosseinmardi, R. Hosseinzadeh, Anticancer
MMP-9/MMP-12 inhibitor on smoke-induced emphysema and airway effect of gallic acid in presence of low level laser irradiation: ROS production
remodelling in Guinea pigs, Thorax 62 (2007) 706e713. and induction of apoptosis and ferroptosis, Canc. Cell Int. 20 (2020) 18.
[60] M.D. Baugh, J. Gavrilovic, I.R. Davies, D.A. Hughes, M.J. Sampson, Monocyte [85] D.K. Maurya, N. Nandakumar, T.P. Devasagayam, Anticancer property of
matrix metalloproteinase production in Type 2 diabetes and controls-a cross gallic acid in A549, a human lung adenocarcinoma cell line, and possible
sectional study, Cardiovasc. Diabetol. 2 (2003) 1e4. mechanisms, J. Clin. Biochem. Nutr. 48 (2011) 85e90.
[61] S. Uemura, H. Matsushita, W. Li, A.J. Glassford, T. Asagami, K.H. Lee, [86] L. Chen, H.Y. Zhang, Cancer preventive mechanisms of the green tea poly-
D.G. Harrison, P.S. Tsao, Diabetes mellitus enhances vascular matrix metal- phenol (-)-epigallocatechin-3-gallate, Molecules 12 (2007) 946e957.
loproteinase activity: role of oxidative stress, Circ. Res. 88 (2001) [87] A.C. Saragusti, M.G. Ortega, J.L. Cabrera, D.A. Estrin, M.A. Marti,
1291e1298. G.A. Chiabrando, Inhibitory effect of quercetin on matrix metalloproteinase 9
[62] I. Ebihara, T. Nakamura, N. Shimada, H. Koide, Increased plasma activity, molecular mechanism and structure-activity relationship of the
metalloproteinase-9 concentrations precede development of micro- flavonoid-enzyme interaction, Eur. J. Pharmacol. 644 (2010) 138e145.
albuminuria in non-insulin-dependent diabetes mellitus, Am. J. Kidney Dis. [88] R. Hoekstra, F.A.L.M. Eskens, J. Verweji, Matrix metalloproteinase inhibitors:
32 (1998) 544e550. current developments and future perspectives, Oncol. 6 (2001) 415e427.
[63] T.T. Nguyen, D. Ding, W.R. Wolter, R.L. Pe rez, M.M. Champion, [89] D.C. Marshall, S.K. Lyman, S. McCauley, M. Kovalenko, R. Spangler, C. Liu,
K.V. Mahasenan, D. Hesek, M. Lee, V.A. Schroeder, J.I. Jones, E. Lastochkin, M. Lee, C. Osullivan, V. Barry-Hamilton, H. Ghermazien, A. Mikels-Vigdal,
M.K. Rose, C.E. Peterson, M.A. Suckow, S. Mobashery, M. Chang, Validation of C.A. Garcia, B. Jorgensen, A.C. Velayo, R. Wang, J.I. Adamkewicz, V. Smith,
matrix metalloproteinase-9 (MMP-9) as a novel target for treatment of Selective allosteric inhibitor of MMP-9 is efficacious in preclinical models of
diabetic foot ulcers in humans and discovery of a potent and selective small- ulcerative colitis and colorectal cancer, PloS One 10 (2015), e0127063.
molecule MMP-9 inhibitor that accelerates healing, J. Med. Chem. 61 (2018) [90] E. Martens, A. Leyssen, I. Van Aelst, P. Fiten, H. Piccard, J. Hu, F.J. Descamps,
8825e8837. P.E. Van den Steen, P. Proost, J. Van Damme, G.M. Liuzzi, P. Riccio, E. Polverini,
[64] J.W. Skiles, N.C. Gonnella, A.Y. Jeng, The design, structure, and therapeutic G. Opdenakker, A monoclonal antibody inhibits gelatinase B/MMP-9 by se-
application of matrix metalloproteinase inhibitors, Curr. Med. Chem. 8 lective binding to part of the catalytic domain and not to the fibronectin or
(2001) 425e474. zinc binding domains, Biochim. Biophys. Acta 1770 (2007) 178e186.
[65] M. Whittaker, C.D. Floyd, P. Brown, A.J.H. Gearing, Design and therapeutic [91] J.F. Pruijt, W.E. Fibbe, L. Laterveer, R.A. Pieters, I.J. Lindley, L. Paemen,
application of matrix metalloproteinase inhibitor, Chem. Rev. 99 (1999) S. Masure, R. Willemze, G. Opdenakker, Prevention of interleukin-8-induced
2735e2776. mobilization of hematopoietic progenitor cells in rhesus monkeys by
[66] R.E. Vandenbroucke, E. Dejonckheere, C. Libert, A therapeutic role for matrix inhibitory antibodies against the metalloproteinase gelatinase B (MMP-9),
metalloproteinase inhibitors in lung diseases? Eur. Respir. 38 (2011) Proc. Natl. Acad. Sci. U. S. A 96 (1999) 10863e10868.
1200e1214. [92] M. Yamamoto, H. Tsujishita, N. Hori, Y. Ohishi, S. Inoue, S. Ikeda, Y. Okada,
[67] A. Kruger, R.E. Kates, D.R. Edwards, Avoiding spam in the proteolytic Inhibition of membrane-type 1 matrix metalloproteinase by hydroxamate
internet: future strategies for anti-metastatic MMP inhibition, Biochim. inhibitors: an examination of the subsite pocket, J. Med. Chem. 41 (1998)
Biophys. Acta 1803 (2010) 95e102. 1209e1217.
[68] E. Maquoi, C. Munaut, A. Colige, C. Lambert, F. Frankenne, A. Noe, F. Grams, [93] M.L. Rothenberg, A.R. Nelson, K.R. Hande, New drugs on the horizon: matrix
H.W. Krell, J.M. Foidart, Stimulation of matrix metalloproteinase-9 expres- metalloproteinase inhibitors, Stem Cell. 17 (1999) 237e240.
sion in human fibrosarcoma cells by synthetic matrix metalloproteinase [94] S. Hanessian, S. Bouzbouz, A. Boudon, G.C. Tucker, D. Peyroulan, Picking the
inhibitors, Exp. Cell Res. 275 (2002) 110e121. S1, S1’ and S2’ pockets of matrix metalloproteinases. A niche for potent
S. Mondal et al. / European Journal of Medicinal Chemistry 194 (2020) 112260 19

acyclic sulfonamide inhibitors, Bioorg. Med. Chem. Lett 9 (1999) 1691e1696. R. Palermo, X-ray structure of a novel matrix metalloproteinase inhibitor
[95] A.M. Chollet, T. Le Diguarher, L. Murray, M. Bertrand, G.C. Tucker, M. Sabatini, complexed to stromelysins, Protein Sci. 10 (2001) 923e926.
A. Pierre, G. Atassi, J. Bonnet, P. Casara, General synthesis of alpha- [116] H.J. Breyholz, M. Schafers, S. Wagner, C. Holtke, A. Faust, H. Rabeneck,
substituted 3-bisaryloxy propionic acid derivatives as specific MMP in- B. Levkau, O. Schober, K. Kopka, C-5-Disubstituted barbiturates as potential
hibitors, Bioorg. Med. Chem. Lett 11 (2001) 295e299. molecular probes for noninvasive matrix metalloproteinase imaging, J. Med.
[96] A.D. Baxter, R. Bhogal, J. Bird, J.F. Keily, D.T. Manallack, J.G. Montana, Chem. 48 (2005) 3400e3409.
D.A. Owen, W.R. Pitt, R.J. Watson, R.E. Wills, Arylsulphonyl hydroxamic acids: [117] J. Wang, C. Medina, M.W. Radomski, J.F. Gilmer, N-Substituted homopiper-
potent and selective matrix metalloproteinase inhibitors, Bioorg. Med. Chem. azine barbiturates as gelatinase inhibitors, Bioorg. Med. Chem. 19 (2011)
Lett 11 (2001) 1465e1468. 4985e4999.
[97] S. Hanessian, N. Moitessier, C. Gauchet, M. Viau, N-Aryl sulfonyl homocys- [118] B. Beutel, J. Song, C.P. Konken, E. Korpos, B. Schinor, H. Gerwien,
teine hydroxamate inhibitors of matrix metalloproteinases: further probing R. Vidyadharan, M. Burmeister, L. Li, G. Haufe, L. Sorokin, New in vivo
of the S(1), S(1)’, and S(2)’ pockets, J. Med. Chem. 44 (2001) 3066e3073. compatible matrix metalloproteinase (MMP)-2 and MMP-9 inhibitors, Bio-
[98] S. Hanessian, D.B. MacKay, N. Moitessier, Design and synthesis of matrix conjugate Chem. 29 (2018) 3715e3725.
metalloproteinase inhibitors guided by molecular modeling. Picking the S(1) [119] J.I. Levin, J.F. DiJoseph, L.M. Killar, A. Sung, T. Walter, M.A. Sharr, C.E. Roth,
pocket using conformationally constrained inhibitors, J. Med. Chem. 44 J.S. Skotnicki, J.D. Albright, The synthesis and biological activity of a novel
(2001) 3074e3082. series of diazepine MMP inhibitors, Bioorg. Med. Chem. Lett 8 (1998)
[99] S. Yamamoto, S. Nakatani, M. Ikura, T. Sugiura, Y. Nishita, S. Itadani, 2657e2662.
K. Ogawa, H. Ohno, K. Takahashi, H. Nakai, M. Toda, Design and synthesis of [120] Y. Tamura, F. Watanabe, T. Nakatani, K. Yasui, M. Fuji, T. Komurasaki,
an orally active matrix metalloproteinase inhibitor, Bioorg. Med. Chem. 14 H. Tsuzuki, R. Maekawa, T. Yoshioka, K. Kawada, K. Sugita, M. Ohtani, Highly
(2006) 6383e6403. selective and orally active inhibitors of type iv collagenase (MMP-9 and
[100] S.M. Yang, R.H. Scannevin, B. Wang, S.L. Burke, Z. Huang, P. Karnachi, MMP-2): n-sulfonylamino acid derivatives, J. Med. Chem. 41 (1998)
L.J. Wilson, K.J. Rhodes, B. Lagu, W.V. Murray, beta-N-Biaryl ether sulfon- 640e649.
amide hydroxamates as potent gelatinase inhibitors: Part 2. Optimization of [121] N.G. Almstead, R.S. Bradley, S. Pikul, B. De, M.G. Natchus, Y.O. Taiwo, F. Gu,
alpha-amino substituents, Bioorg. Med. Chem. Lett 18 (2008) 1140e1145. L.E. Williams, B.A. Hynd, M.J. Janusz, C.M. Dunaway, G.E. Mieling, Design,
[101] J.M. Zapico, P. Serra, J. Garcia-Sanmartin, K. Filipiak, R.J. Carbajo, A.K. Schott, synthesis, and biological evaluation of potent thiazine- and thiazepine-based
A. Pineda-Lucena, A. Martínez, S. Martin-Santamaria, B. de Pascual-Teresa, matrix metalloproteinase inhibitors, J. Med. Chem. 42 (1999) 4547e4562.
A. Ramos, Potent "clicked" MMP2 inhibitors: synthesis, molecular modeling [122] M. Cheng, B. De, S. Pikul, N.G. Almstead, M.G. Natchus, M.V. Anastasio,
and biological exploration, Org. Biomol. Chem. 9 (2011) 4587e4599. S.J. McPhail, C.E. Snider, Y.O. Taiwo, L. Chen, C.M. Dunaway, F. Gu,
[102] B. Fabre, K. Filipiak, J.M. Zapico, N. Diaz, R.J. Carbajo, A.K. Schott, M.E. Dowty, G.E. Mieling, M.J. Janusz, S. Wang-Weigand, Design and syn-
M.P. Martinez-Alcazar, D. Suarez, A. Pineda-Lucena, A. Ramos, B. de Pascual- thesis of piperazine-based matrix metalloproteinase inhibitors, J. Med.
Teresa, Progress towards water-soluble triazole-based selective MMP-2 in- Chem. 43 (2000) 369e380.
hibitors, Org. Biomol. Chem. 11 (2013) 6623e6641. [123] S. Pikul, K. McDow Dunham, N.G. Almstead, B. De, M.G. Natchus, Y.O. Taiwo,
[103] M. Behrends, S. Wagner, K. Kopka, O. Schober, M. Sch€ afers, S. Kumbhar, L.E. Williams, B.A. Hynd, L.C. Hsieh, M.J. Janusz, F. Gu, G.E. Mieling, Hetero-
M. Waller, G. Haufe, New matrix metalloproteinase inhibitors based on g- cycle-based MMP inhibitors with P2’ substituents, Bioorg. Med. Chem. Lett
fluorinated a-aminocarboxylic and a-aminohydroxamic acids, Bioorg. Med. 11 (2001) 1009e1013.
Chem. 23 (2015) 3809e3818. [124] J.I. Levin, J. Chen, M. Du, M. Hogan, S. Kincaid, F.C. Nelson, A.M. Venkatesan,
[104] E. Nuti, A.R. Cantelmo, C. Gallo, A. Bruno, B. Bassani, C. Camodeca, T. Wehr, A. Zask, J. DiJoseph, L.M. Killar, S. Skala, A. Sung, M. Sharr, C. Roth,
T. Tuccinardi, L. Vera, E. Orlandini, S. Nencetti, E.A. Stura, A. Martinelli, G. Jin, R. Cowling, K.M. Mohler, R.A. Black, C.J. March, J.S. Skotnicki, The
V. Dive, A. Albini, A. Rossello, N-O-Isopropyl sulfonamido-based hydrox- discovery of anthranilic acid-based MMP inhibitors. Part 2: SAR of the 5-
amates as matrix metalloproteinase inhibitors: hit selection and in vivo position and P11 groups, Bioorg. Med. Chem. Lett 11 (2001) 2189e2192.
antiangiogenic activity, J. Med. Chem. 58 (2015) 7224e7240. [125] J.I. Levin, J.M. Chen, M.T. Du, F.C. Nelson, T. Wehr, J.F. DiJoseph, L.M. Killar,
[105] J.M. Zapico, A. Puckowska, K. Filipiak, C. Coderch, B. de Pascual-Teresa, S. Skala, A. Sung, M.A. Sharr, C.E. Roth, G. Jin, R. Cowling, L. Di, M. Sherman,
A. Ramos, Design and synthesis of potent hydroxamate inhibitors with Z.B. Xu, C.J. March, K.M. Mohler, R.A. Black, J.S. Skotnicki, The discovery of
increased selectivity within the gelatinase family, Org. Biomol. Chem. 13 anthranilic acid-based MMP inhibitors. Part 3: incorporation of basic amines,
(2015) 142e156. Bioorg. Med. Chem. Lett 11 (2001) 2975e2978.
[106] R. Kiyama, Y. Tamura, F. Watanabe, H. Tsuzuki, M. Ohtani, M. Yodo, Ho- [126] F.C. Nelson, E.D. Santos, J.I. Levin, J.M. Chen, J.S. Skotnicki, J.F. DiJoseph,
mology modeling of gelatinase catalytic domains and docking simulations of M.A. Sharr, A. Sung, L.M. Killar, R. Cowling, G. Jin, C.E. Roth, J.D. Albright,
novel sulfonamide inhibitors, J. Med. Chem. 42 (1999) 1723e1728. Benzodiazepine inhibitors of the MMPs and TACE, bioorg, Med. Chem. Lett.
[107] S.V. Selivanova, T. Stellfeld, T.K. Heinrich, A. Müller, S.D. Kramer, 12 (2002) 2867e2870.
P.A. Schubiger, R. Schibli, S.M. Ametamey, B. Vos, J. Meding, M. Bauser, [127] A. Zask, Y. Gu, J.D. Albright, X. Du, M. Hogan, J.I. Levin, J.M. Chen, L.M. Killar,
J. Hutter, L.M. Dinkelborg, Design, synthesis, and initial evaluation of a high A. Sung, J.F. DiJoseph, M.A. Sharr, C.E. Roth, S. Skala, G. Jin, R. Cowling,
affinity positron emission tomography probe for imaging matrix metal- K.M. Mohler, D. Barone, R. Black, C. March, J.S. Skotnicki, Synthesis and SAR
loproteinases 2 and 9, J. Med. Chem. 56 (2013) 4912e4920. of bicyclic heteroaryl hydroxamic acid MMP and TACE inhibitors, Bioorg.
[108] A.K. Halder, S. Mallick, D. Shikha, A. Saha, D.K. Saha, T. Jha, Design of dual Med. Chem. Lett 13 (2003) 1487e1490.
MMP-2/HDAC-8 inhibitors by pharmacophore mapping, molecular docking, [128] V. Aranapakam, J.M. Davis, G.T. Grosu, J. Baker, J. Ellingboe, A. Zask, J.I. Levin,
synthesis and biological activity, RSC Adv. 5 (2015) 72373e72386. V.P. Sandanayaka, M. Du, J.S. Skotnicki, J.F. DiJoseph, A. Sung, M.A. Sharr,
[109] N. Adhikari, A.K. Halder, S. Mallick, A. Saha, K.D. Saha, T. Jha, Robust design of L.M. Killar, T. Walter, G. Jin, R. Cowling, J. Tillett, W. Zhao, J. McDevitt, Z.B. Xu,
some selective matrix metalloproteinase-2 inhibitors over matrix Synthesis and structure-activity relationship of N-substituted 4-
metalloproteinase-9 through in silico/fragment-based lead identification and arylsulfonylpiperidine-4-hydroxamic acids as novel, orally active matrix
de novo lead modification: syntheses and biological assays, Bioorg. Med. metalloproteinase inhibitors for the treatment of osteoarthritis, J. Med.
Chem. 24 (2016) 4291e4309. Chem. 46 (2003) 2376e2396.
[110] A. Mukherjee, N. Adhikari, T. Jha, A pentanoic acid derivative targeting [129] T. Le Diguarher, A.-M. Chollet, M. Bertrand, P. Hennig, E. Raimbaud,
matrix metalloproteinase-2 (MMP-2) induces apoptosis in a chronic myeloid M. Sabatini, N. Guilbaud, A. Pierre , G.C. Tucker, P. Casara, Stereospecific
leukemia cell line, Eur. J. Med. Chem. 141 (2017) 37e50. synthesis of 5-substituted 2-bisarylthiocyclopentane carboxylic acids as
[111] M.S. Ayoup, M.A. Fouad, H. Abdel-Hamid, S. Ramadan, M.M. Abu-Serie, specific matrix metalloproteinase inhibitors, J. Med. Chem. 46 (2003)
A. Noby, M. Teleb, Battle tactics against MMP-9; discovery of novel non- 3840e3852.
hydroxamate MMP-9 inhibitors endowed with PI3K/AKT signaling attenu- [130] Y.-M. Zhang, X. Fan, D. Chakaravarty, B. Xiang, R.H. Scannevin, Z. Huang,
ation and caspase 3/7 activation via Ugi bis-amide synthesis, Eur. J. Med. J. Ma, S.L. Burke, P. Karnachi, K.J. Rhodes, P.F. Jackson, 1-Hydroxy-2-
Chem. 186 (2020) 111875. pyridinone-based MMP inhibitors: synthesis and biological evaluation for
[112] C.K. Wada, J.H. Holms, M.L. Curtin, Y. Dai, A.S. Florjancic, R.B. Garland, Y. Guo, the treatment of ischemic stroke, Bioorg. Med. Chem. Lett 18 (2008)
H.R. Heyman, J.R. Stacey, D.H. Steinman, D.H. Albert, J.J. Bouska, I.N. Elmore, 409e413.
C.L. Goodfellow, P.A. Marcotte, P. Tapang, D.W. Morgan, M.R. Michaelides, [131] S. Wagner, H.-J. Breyholz, M.P. Law, A. Faust, C. Ho €ltke, S. Schro
€er, G. Haufe,
S.K. Davidsen, Phenoxyphenyl sulfone N-formylhydroxylamines (retro- B. Levkau, O. Schober, M. Scha €fers, K. Kopka, Novel fluorinated derivatives of
hydroxamates) as potent, selective, orally bioavailable matrix metal- the broad-spectrum MMP inhibitors N-hydroxy-2(R)-[[(4-methoxyphenyl)
loproteinase inhibitors, J. Med. Chem. 45 (2002) 219e232. sulfonyl](benzyl)- and (3-picolyl)-amino]-3-methyl-butanamide as potential
[113] F. Grams, H. Brandstetter, S. DAlo, D. Geppert, H.W. Krell, H. Leinert, V. Livi, tools for the molecular imaging of activated MMPs with PET, J. Med. Chem.
E. Menta, A. Oliva, G. Zimmermann, Pyrimidine-2,4,6-triones: a new effective 50 (2007) 5752e5764.
and selective class of matrix metalloproteinase inhibitors, Biol. Chem. 382 [132] E. Nuti, L. Panelli, F. Casalini, S.I. Avramova, E. Orlandini, S. Santamaria,
(2001) 1277e1285. S. Nencetti, T. Tuccinardi, A. Martinelli, G. Cercignani, N. D’Amelio,
[114] H. Brandstetter, F. Grams, D. Glitz, A. Lang, R. Huber, W. Bode, H.W. Krell, A. Maiocchi, F. Uggeri, A. Rossello, Design, synthesis, biological evaluation,
R.A. Engh, The 1.8-A crystal structure of a matrix metalloproteinase 8- and nmr studies of a new series of arylsulfones as selective and potent
barbiturate inhibitor complex reveals a previously unobserved mechanism matrix metalloproteinase-12 inhibitors, J. Med. Chem. 52 (2009) 6347e6361.
for collagenase substrate recognition, J. Biol. Chem. 276 (2001) [133] D.P. Becker, T.E. Barta, L.J. Bedell, T.L. Boehm, B.R. Bond, J. Carroll, C.P. Carron,
17405e17412. G.A. De Crescenzo, A.M. Easton, J.N. Freskos, C.L. Funckes-Shippy, M. Heron,
[115] P. Dunten, U. Kammlott, R. Crowther, W. Levin, L.H. Foley, P. Wang, S. Hockerman, C.P. Howard, J.R. Kiefer, M.H. Li, K.J. Mathis, J.J. McDonald,
20 S. Mondal et al. / European Journal of Medicinal Chemistry 194 (2020) 112260

P.P. Mehta, G.E. Munie, T. Sunyer, C.A. Swearingen, C.I. Villamil, D. Welsch, A combination of docking, QM/MM methods, and MD simulation for binding
J.M. Williams, Y. Yu, J. Yao, Orally Active MMP-1 Sparing a-tetrahydropyranyl affinity estimation of metalloprotein ligands, J. Med. Chem. 48 (2005)
and a-piperidinyl sulfone matrix metalloproteinase (MMP) inhibitors with 5437e5447.
efficacy in cancer, arthritis, and cardiovascular disease, J. Med. Chem. 53 [141] T. Tuccinardi, E. Nuti, G. Ortore, A. Rossello, S.I. Avramova, A. Martinelli,
(2010) 6653e6680. Development of a receptor-based 3D-QSAR study for the analysis of MMP2,
[134] L.J. Wilson, B. Wang, S.-M. Yang, R.H. Scannevin, S.L. Burke, P. Karnachi, MMP3, and MMP9 inhibitors, Bioorg. Med. Chem. 16 (2008) 7749e7758.
K.J. Rhodes, W.V. Murray, Discovery of novel Cobactin-T based matrix met- [142] M. Ramezani, J. Shamsara, A cross-docking study on matrix metal-
alloproteinase inhibitors via a ring closing metathesis strategy, Bioorg. Med. loproteinase family, Antiinflamm. Antiallerg. Agents Med. Chem. 14 (2015)
Chem. Lett 21 (2011) 6485e6490. 164e171.
[135] A. Topai, P. Breccia, F. Minissi, A. Padova, S. Marini, I. Cerbara, In silico [143] Q. Gao, Y. Wang, J. Hou, Q. Yao, J. Zhang, Multiple receptor-ligand based
scaffold evaluation and solid phase approach to identify new gelatinase in- pharmacophore modeling and molecular docking to screen the selective
hibitors, Bioorg. Med. Chem. 20 (2012) 2323e2337. inhibitors of matrix metalloproteinase-9 from natural products, J. Comput.
[136] V. Hugenberg, H.-J. Breyholz, B. Riemann, S. Hermann, O. Schober, Aided Mol. Des. 31 (2017) 625e641.
M. Scha €fers, U. Gangadharmath, V. Mocharla, H. Kolb, J. Walsh, W. Zhang, [144] S. Jana, S.K. Singh, Identification of selective MMP-9 inhibitors through
K. Kopka, S. Wagner, A new class of highly potent matrix metalloproteinase multiple e-pharmacophore, ligand-based pharmacophore, molecular dock-
inhibitors based on triazole-substituted hydroxamates: (radio)synthesis and ing, and density functional theory approaches, J. Biomol. Struct. Dyn. 37
in vitro and first in vivo evaluation, J. Med. Chem. 55 (2012) 4714e4727. (2019) 944e965.
[137] I. Kreituss, E. Rozenberga, J. Zemıtis, P. Trapencieris, N. Romanchikova, [145] J. Hou, Q. Zou, Y. Wang, Q. Gao, W. Yao, Q. Yao, J. Zhang, Screening for the
M. Turks, Discovery of aziridine-triazole conjugates as selective MMP-2 in- selective inhibitors of MMP-9 from natural products based on pharmaco-
hibitors, Chem. Heterocycl. Compd. 49 (2013) 1108e1117. phore modeling and molecular docking in combination with bioassay
[138] J. Hu, M. Yan, C. Pu, J. Wang, P.E. Van den Steen, G. Opdenakker, H. Xu, experiment, hybrid QM/MM calculation, and MD simulation, J. Biomol.
Chemically synthesized matrix metalloproteinase and angiogenesis- Struct. Dyn. 37 (2019) 3135e3149.
inhibiting peptides as anticancer agents, Anti Canc. Agents Med. Chem. 14 [146] N. Adhikari, S.A. Amin, P. Trivedi, T. Jha, B. Ghosh, HDAC3 is a potential
(2014) 483e494. validated target for cancer: an overview on the benzamide-based selective
[139] G. Ledour, G. Moroy, M. Rouffet, E. Bourguet, D. Guillaume, M. Decarme, HDAC3 inhibitors through comparative SAR/QSAR/QAAR approaches, Eur. J.
H. Elmourabit, F. Auge , A.J. Alix, J.Y. Laronze, G. Bellon, W. Hornebeck, J. Sapi, Med. Chem. 157 (2018) 1127e1142.
Introduction of the 4-(4-bromophenyl)benzenesulfonyl group to hydrazide [147] N. Adhikari, S.A. Amin, A. Saha, T. Jha, Exploring in house glutamate in-
analogs of Ilomastat leads to potent gelatinase B (MMP-9) inhibitors with hibitors of matrix metalloproteinase-2 through validated robust chemico-
improved selectivity, Bioorg. Med. Chem. 16 (2008) 8745e8759. biological quantitative approaches, Struct. Chem. 29 (2018) 285e297.
[140] A. Khandelwal, V. Lukacova, D. Comez, D.M. Kroll, S. Raha, S. Balaz,

You might also like