ArticlePDF Available

Pharmacodynamic considerations in the use of matrix metalloproteinase inhibitors in cancer treatment

Authors:

Abstract and Figures

Introduction: Matrix metalloproteinases (MMPs) are classified in the family of zinc-dependent endopeptidases, which can degrade various components of an extracellular matrix and a basement membrane. Studies have demonstrated that MMPs relate to the development of malignant tumors and induce angiogenesis, resulting in the invasion and metastasis of tumor cells. MMPs are highly expressed in malignant tumors and are related to cancer patients’ malignant phenotype and poor prognosis. Therefore, blocking the expression or activity of MMPs may be a promising strategy for cancer treatment. Areas Covered: This study aimed to explain the MMP structure, regulatory mechanism, and carcinogenic effect; investigate the matrix metalloproteinase-inhibitors (MMPIs) that are currently used in clinical trials for cancer treatment; and summarize the trial results. Expert Opinion: Currently, the results of clinical trials that have used MMPIs as anticancer agents are unsatisfactory. However, MMPs remain an attractive target for cancer treatment. For example, development of the specific peptide or antibodies in targeting the hemopexin domain of MMP-2 may be a new therapeutic direction. The design and development of MMPIs that have selectivity will be the primary focus in future studies.
Content may be subject to copyright.
!"##$%&'()$*$+,-./0/,-)$,1$233&))$2-.$")&$32-$4&$1,"-.$20
5006788999:02-.1,-#/-&:3,(8230/,-8;,"'-2#<-1,'(20/,-=;,"'-2#+,.&>/&(0?@
!"#$%"&'()*+$A<-)0/0"0&$,1$B,1092'&C !&,-+$DE$!&4'"2'F$?@DGH$I07$@@7EJ
./0-1,(203$3"$("$(!145(6-,&)"%378(9(:"/3;"%"5*
<==>+(?@ABCDBDD(EF13$,G(?@AAC@HI@(E2$%3$-G(J"41$&%(K"8-0&5-+(K,,0+LL###M,&$'N"$%3$-M;"8L%"3L3-8,BI
FK&18&;"'*$&83;(;"$73'-1&,3"$7(3$(,K-(47-("N
8&,13/(8-,&%%"01",-3$&7-(3$K3)3,"17(3$(;&$;-1
,1-&,8-$,
J3&C=3$(O&$5P(QK3&"CR-$(S3$P(=K3KCQK3(=4(9(=K4$CT&(O&$5
:"(;3,-(,K37(&1,3;%-+$K/2LB/-$M2-NH$+5/2,LO&-$P/-H$B5/5L+5/$B"$*$B5"-L!2$M2-N$Q?@DGR
S52'(23,.F-2(/3$3,-)/.&'20/,-)$/-$05&$")&$,1$(20'/T$(&02##,6',0&/-2)&$/-5/4/0,')$/-
32-3&'$0'&20(&-0H$UT6&'0$V6/-/,-$,-$W'"N$X&024,#/)($*$%,T/3,#,NFH$D?7?H$DYDL?@@H$WV<7
D@:DEDZ8DZ[?E?EE:?@DG:DD\DJ?@
:"(%3$U(,"(,K37(&1,3;%-+($5006788.T:.,/:,'N8D@:DEDZ8DZ[?E?EE:?@DG:DD\DJ?@
I33&60&.$2"05,'$]&')/,-$6,)0&.$,-#/-&7$D?
W&3$?@DE:
S"4#/)5&.$,-#/-&7$DD$K2-$?@DG:
B"4(/0$F,"'$2'0/3#&$0,$05/)$;,"'-2#$
I'0/3#&$]/&9)7$DD
^/&9$'&#20&.$2'0/3#&)$
^/&9$+',))(2'_$.202
REVIEW
Pharmacodynamic considerations in the use of matrix metalloproteinase
inhibitors in cancer treatment
Jia-Sin Yang
a,b
, Chiao-Wen Lin
c,d
, Shih-Chi Su
e,f
and Shun-Fa Yang
a,b
a
Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan;
b
Institute of Medicine, Chung Shan Medical
University, Taichung, Taiwan;
c
Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan;
d
Department of Dentistry,
Chung Shan Medical University Hospital, Taichung, Taiwan;
e
Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung
Memorial Hospital, Keelung, Taiwan;
f
Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung
Memorial Hospitals, Linkou, Taiwan
ABSTRACT
Introduction: Matrix metalloproteinases (MMPs) are classified in the family of zinc-dependent
endopeptidases, which can degrade various components of an extracellular matrix and a base-
ment membrane. Studies have demonstrated that MMPs relate to the development of malignant
tumors and induce angiogenesis, resulting in the invasion and metastasis of tumor cells. MMPs
are highly expressed in malignant tumors and are related to cancer patientsmalignant pheno-
type and poor prognosis. Therefore, blocking the expression or activity of MMPs may be a
promising strategy for cancer treatment.
Areas Covered: This study aimed to explain the MMP structure, regulatory mechanism, and
carcinogenic effect; investigate the matrix metalloproteinase-inhibitors (MMPIs) that are currently
used in clinical trials for cancer treatment; and summarize the trial results.
Expert Opinion: Currently, the results of clinical trials that have used MMPIs as anticancer agents
are unsatisfactory. However, MMPs remain an attractive target for cancer treatment. For example,
development of the specific peptide or antibodies in targeting the hemopexin domain of MMP-2
may be a new therapeutic direction. The design and development of MMPIs that have selectivity
will be the primary focus in future studies.
ARTICLE HISTORY
Received 29 September 2015
Accepted 10 December 2015
Published online
8 January 2016
KEYWORDS
Cancer; matrix
metalloproteinases; matrix
metalloproteinases inhibitor;
pharmacodynamics
1. Introduction
Cancer has been detected for years and remains a
major threat to the health of human beings worldwide.
According to GLOBOCAN 2012, the global incidence of
cancer was 14,067,894 persons, and the global mortality
was 8,201,575 persons.[1] The United States estimated
that 1,658,370 new cancer cases and 589,430 deaths
caused by cancer would occur in 2015.[2] Despite the
advanced medical technology that renders cancer a
curable disease, the mortality rate of cancer remains
high. Cancer has the following characteristics: (1) sus-
taining proliferative signaling, (2) evading growth sup-
pressors, (3) resisting cell death, (4) enabling replicative
immortality, (5) inducing angiogenesis, and (6) activat-
ing invasion and metastasis.[3] Cancer metastasis and
invasion are crucial causes of the high mortality rate
and relate to the high expression of matrix metallopro-
teinases (MMPs).[46] MMPs are a group of proteinases,
which degrade and remodel extracellular matrix (ECM).
However, the high expression of MMPs is commonly
observed in various malignant tumors. In addition, the
MMP expression in the metastasis tissues is higher than
that in the primary tumor tissues.[79] Therefore, MMPs
play a vital role during the progression and metastasis
of malignant tumors. Designing and developing MMP
inhibitors (MMPIs) as anticancer agents is feasible for
cancer treatment.[1012] Thus, this study discussed the
rationality of developing targeted therapy against
MMPs and summarized the use of MMPIs in clinical
trials and the results of these trials.
2. Matrix metalloproteinases
MMPs were first found in 1962 in the metamorphosis of
a tadpole tail. In 1968, MMPs were purified from human
skin. MMPs consist of zinc-dependent endopeptidases
and are synthesized from connective tissue cells and
proinflammatory cells, such as fibroblasts, osteoblasts,
endothelial cells, macrophages, neutrophils, and lym-
phocytes. Most MMPs are secreted in the form of unac-
tivated zymogen (pro-MMPs). The bait region on the
CONTACT Shun-Fa Yang ysf@csmu.edu.tw; Shih-Chi Su ssu1@cgmh.org.tw Institute of Medicine, Chung Shan Medical University, 110 Chien-Kuo
N. Road, Section 1, Taichung 402, Taiwan
EXPERT OPINION ON DRUG METABOLISM & TOXICOLOGY, 2016
VOL. 12, NO. 2, 191200
http://dx.doi.org/10.1517/17425255.2016.1131820
© 2016 Taylor & Francis
Downloaded by [Institute of Software] at 00:58 15 February 2016
propeptide of MMPs is removed through hydrolysis by
serine proteases, furin, plasmin, or other MMP proteins.
Therefore, the MMPs are unstable. However, the MT-
MMPs are active and can, in turn, activate MMP-2. In
vitro investigations revealed that pro-MMP-2 (72 kDa)
activation requires contributions from MT1-MMP and
tissue inhibitor of metalloproteinase-2 (TIMP-2).[13]
Active MT1-MMP anchored to the cell surface acts as a
receptor for TIMP-2, which binds to the active site of
MT1-MMP. Subsequently, the interaction between the
cysteine on the propeptide and the Zn
2+
ions on the
catalytic domain is destroyed through a cysteine switch
mechanism, thus activating the MMPs.[14]
MMPs are typically classified into collagenases, gela-
tinases, stromelysins, and membrane-type MMPs (MT-
MMPs) according to the specificity of their substrates
and their location in cells. However, this classification
method is unsuitable for some MMPs. Therefore, the
MMPs are newly classified into archetypal MMPs, matri-
lysins, gelatinases, and furin-activatable MMPs accord-
ing to the domain organization of MMPs (Table 1).[15]
The basic structure of the archetypal MMPs involves a
signal peptide (approximately 20 amino acids), a pro-
peptide domain (approximately 80 amino acids) with a
cysteine-switch motif, a Zn
2+
-containing catalytic
domain (approximately 170 amino acids), and a hemo-
pexin domain (approximately 200 amino acids). The
hinge region (a linker peptide of variable length) is
connected to the catalytic domain. The archetypal
MMPs can be further divided into collagenases, strome-
lysins, and otheraccording to the specificity of their
substrates. Collagenases include MMP-1 (collagenases-
1), MMP-8 (collagenases-2), and MMP-13 (collagenases-
3) and can lyse a collagen triple helix. Stromelysins
comprise MMP-3 (stromelysin-1) and MMP-10 (strome-
lysin-2) and have the same structure as that of collage-
nases. Stromelysins can degrade numerous ECM
components and activate procollagenases [16] and
proMMP-9 [17] but cannot degrade native collagen.
Other archetypal MMPs encompass MMP-12 (metalloe-
lastase), MMP-19, MMP-20 (enamelysin), and MMP-27,
which cannot be classified into collagenases or strome-
lysins because their sequence and substrate specificity
are different from that of the other two. Matrilysins
consist of MMP-7 (matrilysins-1) and MMP-26 (matrily-
sins-2) but lack the carboxy-terminal hemopexin
domain.[18] Gelatinases, involving MMP-2 (gelatinase-
A) and MMP-9 (gelatinase-B), have a fibronectin type II
motif in the catalytic domain and thus bind and
degrade denatured collagen or gelatin.[19] Furin-
activatable MMPs contain a furin recognition motif
between propeptide and the catalytic domain and can
be activated in cells through furin-like proteases. Furin-
activatable MMPs are classified into secreted MMPs, MT-
MMPs, and unusual type II transmembrane MMPs.
Secreted MMPs contain MMP-11 (stromelysin-3), MMP-
21, and MMP-28 (epilysin), which are secreted in an
activated form. MT-MMPs encompass membrane-
anchoring domains; therefore, they are located on the
cell surface to control the environment surrounding the
cells. According to the types of membranes to which
they adhere, MT-MMPs are divided into type I trans-
membrane MT-MMPs (involving MMP-14 [MT1-MMP],
MMP-15 [MT2-MMP], MMP-16 [MT3-MMP], and MMP-
24 [MT5-MMP]) and glycosylphosphatidylinositol MT-
MMPs (containing MMP-17 [MT4-MMP] and MMP-25
[MT6-MMP]).[20] Type II transmembrane MMPs contain
MMP-23A and MMP-23B, which have the same amino
acid sequence but originate from different human
genes. Type II transmembrane MMPs lack signal pep-
tide, cysteine-switch motif, or hemopexin domain but
contain a C-terminal tail with cysteine-array and immu-
noglobulin domains, and an N-terminal with type II
transmembrane domain. The activated MMPs generally
can degrade all the ECM and basement-membrane
components, including aggrecan, collagen, elastin,
fibronectin, gelatin, and laminin.[21] MMPs are indis-
pensable in embryonic development, angiogenesis,
cell growth, wound healing, and other physiological
development processes that require tissue remodeling.
[22,23] However, from a pathological perspective,
MMPs may facilitate the invasion of tumor cells into
connective tissues and blood vessel walls, resulting in
tumor metastasis.[2426]
The activity of MMPs is controlled through the
mechanisms of gene expression, compartmentalization,
zymogen activation, and enzyme inhibition. The endo-
genous inhibitor of MMPs, namely α2-macroglobulin,
and the tissue inhibitors of metalloproteinases (TIMPs)
are primarily responsible for inhibiting MMPs [27,28]
and can also participate in MMP activation.[13] The
α2-macroglobulin is a large serum albumin, which
Article highlights.
MMPs are classified in the family of zinc-dependent endopepti-
dases, which can degrade various components of an ECM and a
basement membrane.
MMPs are highly expressed in malignant tumors and are related
to cancer patientsmalignant phenotype and poor prognosis.
Blocking the expression or activity of MMPs may be a promising
strategy for cancer treatment.
MMPIs can be divided into four types: peptidomimetics, non-
peptidomimetics, tetracycline-like derivatives, and BPs.
The failed application of MMPIs to cancer treatment may result
from their lack of selectivity for MMPs. Therefore, developing
MMPIs that have selectivity is a new direction for cancer treat-
ment.
This box summarizes key points contained in the article.
192 J.-S. YANG ET AL.
Downloaded by [Institute of Software] at 00:58 15 February 2016
Table 1. MMP family.
Domain composition
Enzyme group MMP
Chromosomal
location Substrates SP Pro CS RX[R/K]R Cat FNII LK1 Hpx LK2 TM GPI Cyt CyR-Ig
Archetypal MMPs
Collagenases
Collagenases-1 MMP-1 11q22.222.3 Collagen I, II, III, VII, VIII, X, XI, casein, perlecan,
entactin, laminin, proMMP-1, 2, 9, serpins
+ + + +++
Collagenases-2 MMP-8 11q22.222.3 Collagen I, II, III, VII, VIII, X, entactin, gelatin + + + +++
Collagenases-3 MMP-13 11q22.222.3 Collagen I, II, III, VII, X, XVIII, gelatin, entactin,
tenascin, aggregan
+ + + +++
Stromelysins
Stromelysin-1 MMP-3 11q22.222.3 Laminin, aggregan, gelatin, fibronectin + + + +++
Stromelysin-2 MMP-10 11q22.222.3 Collagen I, III, IV, gelatin, elastin, proMMP-1, 8,
10
+ + + +++
Other
Metalloelastase MMP-12 11q22.222.3 Elastin, gelatin, collagen I, IV, fibronectin,
laminin, vitronectin, proteoglycan
+ + + +++
Stromelysin-4 (RASI-1) MMP-19 12q14 Collagen I, IV, tenascin, gelatin, laminin + + + +++
Enamelysin MMP-20 11q22 Laminin, amelogenin, aggregan + + + +++
C-MMP MMP-27 11q24 + + + +++
Matrilysins
Matrilysins-1 MMP-7 11q22.222.3 Collagen I, IV, V, IX, X, XI, XVIII, fibronectin,
laminin, gelatin, aggregan, proMMP-9
+ + + +
Matrilysins-2 (endometase) MMP-26 11q22.2 Gelatin, collagen IV, proMMP-9 + + + +
Gelatinases
Gelatinase-A MMP-2 16q13 Gelatin, fibronectin, elastin, laminin, collagen
I, III, IV, V, VII, X, XI, vitronectin, decorin,
plasminogen
+ + + + + + +
Gelatinase-B MMP-9 20q11.2q13.1 Gelatin, collagen I, IV, V, VII, X, XI, XVIII,
vitronectin, elastin, laminin, fibronectin,
proMMP-2, 9
+ + + + + + +
Furin-activatable MMPs
Secreted
Stromelysin-3 MMP-11 22q11.2 Fibronectin, laminin, aggregan, gelatin + + + + + ++
Epilysin MMP-28 17q11.2 Casein + + + + + ++
Type I transmembrane
MT1-MMP MMP-14 14q12.2 Collagen I, II, III, aggregan, laminin, gelatin,
proMMP-2, 13
+ + + + + + + + + +
MT2-MMP MMP-15 16q12.2 Proteoglycans, proMMP-2 + + + + + + + + + +
MT3-MMP MMP-16 8q21 Collagen III, fibronectin, proMMP-2 + + + + + + + + + +
MT5-MMP MMP-24 20q11.2 Fibrinogen, gelatin, proMMP-2 + + + + + + + + + +
GPI-anchored
MT4-MMP MMP-17 12q24 Gelatin, fibrinogen, proMMP-2 + + + + + ++++
MT6-MMP (Leukolysin) MMP-25 16q13.3 Collagen IV, gelatin, proMMP-2, 9 + + + + + ++++
Type II transmembrane
CA-MMP MMP-23 1p36.3 Gelatin ++++ +
MMP: Matrix metalloproteinase; SP: Signal peptide; Pro: Propeptide domain; CS: Cysteine-switch motif; RX[R/K]R: Proprotein convertase recognition sequence; Cat: Catalytic domain; FNII: Fibronectin type II motif; LK:
Linker; Hpx: Hemopexin domain; TM: Transmembrane domain; GPI: Glycosylphophatidylinositol anchor; Cyt: Cytoplasmic tail domain; CyR-Ig: Cysteine-rich and immunoglobulin domain.
EXPERT OPINION ON DRUG METABO LISM & TOXICOLOGY 193
Downloaded by [Institute of Software] at 00:58 15 February 2016
partially regulates the activity of MMPs. It entraps MMPs
to prevent them from accessing substrates.[29] TIMPs
contain proteins related to TIMP-1, TIMP-2, TIMP-3, and
TIMP-4.[28,30] TIMPs are responsible for postactivation
inhibition of MMPs; they also participate in MMP activa-
tion, and regulate fibrosis and inflammation. Generally,
all TIMPs are capable of inhibiting all known MMPs;
however, the efficacy of MMP inhibition varies with
each TIMP. For example, TIMP-1 is a poor inhibitor of
MT1-MMP, MT3-MMP, MT5-MMP, and MMP-19. TIMP-2
is unique in that, in addition to inhibiting MMP activity,
it selectively interacts with MT1-MMP to facilitate the
cell-surface activation of proMMP-2. Moreover, TIMPs
and the active zinc-binding site of MMPs together
form noncovalent stoichiometric complexes to inhibit
the enzyme activity of MMPs.[31,32] MMPs play a criti-
cal role in the development of malignant tumors. When
the balance between TIMPs and MMPs is disturbed,
causing the MMP activity to increase, tissue degrada-
tion may occur in pathological states.[33,34] Thus, inhi-
biting MMPs can be a direction for cancer treatment.
Although the endogenous TIMPs can inhibit the patho-
logical activity of MMPs,[35] they are broad-spectrum
inhibitors and lack selectivity for individual MMPs. In
addition, TIMPs are involved in a number of physiolo-
gical processes that are not related to MMPs.[3638]
Therefore, developing other MMPIs is vital.
3. Application of matrix metalloproteinase
inhibitors to cancer treatment
The importance of MMPs in cancer progression is
widely known. In the past 50 years, the medical field
has emphasized developing new MMPIs and investigat-
ing their application to cancer treatment. MMPIs can be
divided into four types: peptidomimetics, nonpeptido-
mimetics, tetracycline-like derivatives, and bisphospho-
nates (BPs). This study aimed to investigate the basic
and clinical application of the four types of MMPIs to
cancer treatment and the outcomes of such application
(Table 2).
3.1. Peptidomimetic matrix metalloproteinase
inhibitors
Peptidomimetic MMPIs are pseudopeptide derivatives
that simulate the active structure of MMPs.[39] They
reversibly bind to the active site of MMPs to chelate
zinc atoms.[40] Some zinc-binding groups, such as ami-
nocarboxylates, carboxylates, hydroxamates, sulfhy-
dryls, and derivatives of phosphoric acid, have been
tested for their effect on inhibiting MMP activity.
3.1.1 Hydroxamate derivatives
Hydroxamate derivatives, such as batimastat, marima-
stat, and solimastat, are the inhibitors most commonly
used in clinical practices. These hydroxamate inhibitors
are small peptide analogs of fibrillar collagens, which
inhibit MMP activity by interacting with Zn
2+
ions
located at the catalytic site of MMPs.
3.1.1.1 Batimastat (BB-94). Batimastat is a nonorally
bioavailable low-molecular-weight hydroxamate-based
inhibitor, which was the first MMPI used for clinical
trials.[41] Through directly binding to the Zn
2+
ions on
the MMP active site, batimastat inhibits the activity of
Table 2. Structure and MMP inhibition of synthetic MMPIs.
Class Compound Structure
Specificity
of the
inhibition
Peptidomimetic
MMPIs
Batimastat
(BB-94)
MMP-1, 2,
7, 9
Marimastat
(BB-2516)
MMP-1, 2,
3, 7, 9, 12
Solimastat
(BB-3644)
Broad-
spectrum
Nonpeptidomimetic
MMPIs
Prinomastat
(AG 3340)
MMP-2, 3,
7, 9, 13, 14
Tanomastat
(BAY
129566)
MMP-2, 3,
9, 13, 14
Rebimastat
(BMS-
275291)
MMP-1, 2,
3, 7, 9, 14
MMI-270
(CGS27023A)
MMP-1, 2, 3
Tetracycline
derivatives
Doxycycline MMP-2, 9
Metastat
(CMT-3; Col-
3)
MMP-1, 2,
8, 9, 13
Bisphosphonates Zoledronic
acid
(Zoledronate)
MMP-2, 9
MMP: Matrix metalloproteinase; MMPIs: Matrix metalloproteinase inhibitors.
194 J.-S. YANG ET AL.
Downloaded by [Institute of Software] at 00:58 15 February 2016
MMP-1, MMP-2, MMP-7, and MMP-9.[42] Batimastat
inhibits the growth of various cancer cell lines and has
no cytotoxicity in vitro.[43] Some in vivo studies have
verified the antitumor effect of batimastat in animal
models.[4452] However, batimastat did not yield satis-
factory reactions in the phase I and II clinical trials, and
the trial was halted in phase III.[53] Because of its poor
solubility and low oral bioavailability, batimastat can
only be administered through intrapleural and intraper-
itoneal injection to assess cancer patients participating
in clinical trials.[41,54,55] However, this method has not
been widely accepted in cancer treatment.
3.1.1.2. Marimastat (BB-2516). Marimastat is a syn-
thetic MMPI with a low molecular weight and an orally
bioavailable MMPI that was first used for clinical trials
and can inhibit the activity of MMP-1, MMP-2, MMP-3,
MMP-7, MMP-9, and MMP-12. In the experimental
metastasis models of lung and breast cancer, marima-
stat reduced the number and size of the metastatic
lesions of the animals in the experimental and control
groups.[56] In clinical trials, marimastat exhibited ade-
quate efficacy in delaying disease progression.
However, because of limited dosage, the significance
of toxicity could not be determined. Patients using
marimastat experienced substantial musculoskeletal
pain and inflammation. Because no statistically signifi-
cant difference was observed between the trial results
regarding symptom deterioration and the overall survi-
val rate, the development of marimastat has been dis-
continued.[57]
3.1.1.3. Solimastat (BB-3644). Solimastat is an oral,
broad-spectrum MMPI; its structure is similar to that of
marimastat and batimastat. Currently, the inhibitory
activity of solimastat is still unknown. Although solima-
stat expresses anticancer characteristics similar to that
of marimastat, the phase I clinical trial that adminis-
tered solimastat to solid cancer failed.[58] Currently,
no further development and application of peptidomi-
metic MMPIs for cancer treatment have been
undertaken.
3.2. Nonpeptidomimetic matrix metalloproteinase
inhibitors
When used clinically, peptidomimetic MMPIs exhibited
problems of poor oral bioavailability (except for mari-
mastat) and lack of MMP specificity. To improve the
development of these drugs, some nonpeptidomimetic
MMPIs were synthesized according to the 3D X-ray
crystallography of the MMP active sites. Currently, the
nonpeptidomimetic MMPIs administered to cancer
patients for clinical assessment involve prinomastat
(AG 3340), tanomastat (BAY 129566), rebimastat
(BMS-275291), and MMI-270 (CGS27023A), which fea-
ture higher specificity and oral bioavailability than the
peptidomimetic MMPIs do.
3.2.1. Prinomastat (AG 3340)
Prinomastat is a synthetic, nonpeptidic collagen-
mimicking MMPI with a low molecular weight, which
can inhibit the activity of MMP-2, MMP-3, MMP-7, MMP-
9, MMP-13, and MMP-14. Studies using xenograft mod-
els have demonstrated the antitumor effect of prinoma-
stat.[5964] Moreover, in a phase I and pharmacokinetic
study of prinomastat, Hande et al. found that prinoma-
stat was rapidly absorbed, with peak concentration
occurring within the first hour after dosing.[65]
However, in phase III trials, the application of prinoma-
stat combined with chemotherapy to the treatment of
advanced non-small-cell lung cancer failed to improve
the treatment efficacy.[66]
3.2.2. Tanomastat (BAY 129566)
Tanomastat is an orally bioavailable biphenyl synthetic
MMPI that can inhibit the activity of MMP-2, MMP-3,
MMP-9, MMP-13, and MMP-14.[67] An in vivo experi-
ment demonstrated that tanomastat can inhibit angio-
genesis.[68] A phase I and pharmacokinetic study of
tanomastat was tested, following 60 mg/m
2
infusion,
the mean peak plasma etoposide concentration was
11.7 ± 2.8 mg/ml.[69] Although tanomastat exhibited
satisfactory tolerance and generated no musculoskele-
tal side effects, it exhibited no reactions in phase I
clinical trials [70] and had no influence on the progres-
sion-free survival or overall survival rate in phase III
clinical trials.[71] The clinical development of tanoma-
stat has been terminated.
3.2.3. Rebimastat (BMS-275291)
Rebimastat is an orally bioavailable MMPI containing
thiol zinc-binding groups, which can inhibit the activity
of MMP-1, MMP-2, MMP-3, MMP-7, MMP-9, and MMP-
14.[64] In a preclinical study, rebimastat inhibited tumor
progression in rodent tumor models.[72] Additionally,
in a phase I study, rebimastat did not exhibit dose-
limiting joint toxicity and no dose-limiting toxicities
occurred at 900, 1800, or 2400 mg/day.[73] The study
also found that the mean plasma concentration of rebi-
mastat at trough exceeded the calculated in vitro IC(80)
value for MMP-2 and IC(90) value for MMP-9 at the
recommended phase II dose of 1200 mg/day.[73]
However, rebimastat displayed an adverse effect in a
phase II trial on early stage breast cancer and a phase III
trial on non-small-cell lung carcinoma.[74,75]
EXPERT OPINION ON DRUG METABO LISM & TOXICOLOGY 195
Downloaded by [Institute of Software] at 00:58 15 February 2016
3.2.4. MMI-270 (CGS27023A)
MMI-270 is a broad-spectrum nonpeptidic hydroxamic
acid derivative MMPI that can inhibit MMP-1, MMP-2,
and MMP-3. In an in vitro experiment, MMI-270 could
not inhibit the proliferation of cancer cell lines but
could inhibit the invasion of tumor cells into Matrigel
and the angiogenesis of various preclinical models. In a
phase I clinical trial, the effect of MMI-270 on stabilizing
the disease progression was unsatisfactory. Moreover,
when a high dose of MMI-270 was administered, a
widespread maculopapular rash and musculoskeletal
side effects were observed.[76] Moreover, Eatock et al.
found that 5-fluorouracil (5-FU)/folinic acid with MMI-
270 at a dose of 300 mg twice daily is well tolerated;
however, MMI-270 has no significant effect on 5-FU
pharmacokinetics.[77] In a clinical trial on the treatment
of non-small-cell lung carcinoma, the administration of
MMI-270 was terminated because of its poor tolerance
and the side effect of joint and muscle pain.[78]
3.3. Tetracycline derivatives
Tetracycline derivatives inhibit not only MMP activity
but also MMP production. Specifically, by chelating
with zinc atoms at the MMP binding site, tetracycline
blocks the activity of mature MMPs, interferes with the
hydrolysis and activation of pro-MMP proteins, reduces
MMP expression, and protects MMPs from proteolytic
and oxidative degradation.[79,80] The family of tetracy-
cline derivatives involves classic tetracycline antibiotics,
such as doxycycline, and newer tetracycline analogs
(that have been chemically modified to eliminate their
antimicrobial activity), such as Col-3 (metastat).
3.3.1. Doxycycline
Doxycycline is a type of classic antimicrobial tetracy-
cline and has been widely studied as an anticancer
agent.[81,82] Doxycycline inhibits the secretion of
MMP-2 and MMP-9 and noncompetitively inhibits the
activity of MMP-2 and MMP-9 in breast carcinoma cells.
[83] In in vitro studies, doxycycline has inhibited the
proliferation of cancer cells and induced cell apoptosis,
reducing their ability to invade and metastasize.[81,83
87] In an in vivo study, doxycycline combined with
batimastat enhanced the inhibition of tumor metasta-
sis.[46] However, in a phase II clinical trial on treating
renal cell carcinoma, the administration of doxycycline
and interferon-αdid not improve the clinical result.[88]
3.3.2. Metastat (CMT-3; Col-3)
Metastat is one of the chemically modified tetracyclines
which can inhibit the activity of MMP-1, MMP-2, MMP-8,
MMP-9, and MMP-13. In the metastatic prostate cancer
model, metastat inhibited cell proliferation, invasion,
tumor growth, and metastasis.[85,89] In a phase I trial,
metastat exerted an antitumor effect on the soft tissue
sarcoma.[90] Moreover, in the pharmacokinetic study of
metastat, Syed et al. found that the values for median
time at which peak plasma concentration is achieved
and ranges on both days 1 and 29 were identical.[90]
However, in the phase II trial, no objective responses
were observed in patients with advanced and/or meta-
static soft tissue sarcoma.[91]
3.4. Bisphosphonates
BPs, also known as diphosphonates, are used to prevent
bone mass loss and treat osteoporosis. After being used
for several years on human beings, BPs have been verified
as having low toxicity and high tolerance and have exhib-
ited the potential to be used in treating MMP-related
human diseases. BPs can inhibit MMP-1, MMP-2, MMP-3,
MMP-7, MMP-8, MMP-9, MMP-12, MMP-13, and MMP-14.
Additionally, they can inhibit TIMP-2 from degrading
MMP-2 and enhance the inactivation of MMP-2.[92] The
degree of tumor cell invasion can also be reduced.[93]
Recent studies have reported that BP compounds and
zoledronic acid (zoledronate) can inhibit the growth,
migration, and invasion of breast cancer cells [94] and
improve outcomes for breast cancer patients.[95]
4. Conclusion
Although many valuable studies have explored the role
of MMPs in cancer, the regulatory role of MMPs has not
been fully clarified. Despite the incomplete data, MMPs
can be used as the auxiliary marker for diagnosing
cancer metastasis. MMPs are the new therapeutic tar-
gets of cancer; developing drugs that can inhibit the
generation and activity of MMPs can contribute to inhi-
biting cancer progression and enhancing treatment
efficacy. Numerous MMPIs have been applied to clinical
trials to verify their efficacy for cancer treatment.
However, no promising targeted drug therapies have
been used to inhibit MMPs for cancer treatment.
Further research is required to explore the potential
development of MMPIs and their effects on disease
diagnosis and treatment.
5. Expert opinion
Studies on conventional anticancer drugs have empha-
sized how cancer cells are killed by chemical com-
pounds. However, metastasis is one of the primary
factors resulting in patient death.[3] Numerous studies
196 J.-S. YANG ET AL.
Downloaded by [Institute of Software] at 00:58 15 February 2016
have reported that MMPs expedite cancer progression
by increasing the growth, migration, invasion, metasta-
sis, and angiogenesis of cancer cells. In normal organ-
isms, a dynamic balance between MMPs and TIMPs (i.e.
the endogenous inhibitor for MMPs) is maintained to
ensure the normal functioning of the organisms.
However, such balance may be destroyed by cancer,
resulting in the high expression of MMPs. Thus, inhibit-
ing the activity of MMPs is a reasonable method of
cancer treatment.
Most MMPIs have produced promising results in
preclinical studies; however, no significant results have
been observed in current clinical trials. This may have
occurred for several reasons. For example, MMP biology
is different between human beings and experimental
animals, inhibiting MMPs is more effective in metastasis
prevention than in reducing tumor size, drug dosage is
determined according to the severity of side effects
instead of the inhibition of tissue penetration or cataly-
tic activity, and the specific expression of MMPs in
tumors was still unclear at the trial design stage.[96]
Moreover, because the catalytic sites of most MMPs
have high homology, MMPIs lack specificity and, there-
fore, cannot be specifically applied to various types of
MMPs. The nonspecific binding effect may cause an
MMP to be bound to other MMPs necessary to the
organisms. Because a variety of MMPs are diluted, the
effective dosage for the human body may be higher
than expected. Clinically, the failed application of
MMPIs to cancer treatment may result from their lack
of selectivity for MMPs. Therefore, developing MMPIs
that have selectivity is a new direction for cancer treat-
ment. For example, the peptides or blocking antibodies
that can modulate exosite-mediated cell surface inter-
actions and functions in many potential alternative
enzyme domains may present as targets for designing
highly selective MMPIs.[7] Moreover, in contrast to all
other secreted MMPs, pro-MMP-9 and pro-MMP-2 bind
to TIMPs through their hemopexin domain.[97,98]
Those studies suggest that inhibition either substrate
binding or proenzyme activation of MMPs may all
potentially applied for MMPs targeting therapy. The
hemopexin domain of MMP-2 and MMP-9 may be a
viable target to specific abrogates their functions.
Development of the specific peptide or antibodies in
targeting the hemopexin domain of MMPs may be a
new therapeutic direction in targeting cancers.
Declaration of Interest
The author has no relevant affiliations or financial involve-
ment with any organization or entity with a financial interest
in or financial conflict with the subject matter or materials
discussed in the manuscript. This includes employment, con-
sultancies, honoraria, stock ownership or options, expert tes-
timony, grants or patents received or pending, or royalties.
References
Papers of special note have been highlighted as either of
interest () or of considerable interest (••) to readers.
1. Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer inci-
dence and mortality worldwide: sources, methods and
major patterns in GLOBOCAN 2012. Int J Cancer.
2015;136:E359386.
2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA
Cancer J Clin. 2015;65:529.
3. Hanahan D, Weinberg RA. Hallmarks of cancer: the next
generation. Cell. 2011;144:646674.
4. Brown GT, Murray GI. Current mechanistic insights into
the roles of matrix metalloproteinases in tumour invasion
and metastasis. J Pathol. 2015;237:273281.
5. Bauvois B. New facets of matrix metalloproteinases MMP-
2 and MMP-9 as cell surface transducers: outside-in sig-
naling and relationship to tumor progression. Biochim
Biophys Acta. 2012;1825:2936.
6. Sampieri CL, Leon-Cordoba K, Remes-Troche JM. Matrix
metalloproteinases and their tissue inhibitors in gastric
cancer as molecular markers. J Cancer Res Ther.
2013;9:356363.
7. Chien MH, Lin CW, Cheng CW, et al. Matrix metallopro-
teinase-2 as a target for head and neck cancer therapy.
Expert Opin Ther Targets. 2013;17:203216.
•• This article gives an overview of MMP-2 in cancer
therapy.
8. Hsin CH, Chen MK, Tang CH, et al. High level of plasma
matrix metalloproteinase-11 is associated with clinico-
pathological characteristics in patients with oral squa-
mous cell carcinoma. PLoS One. 2014;9:e113129.
9. Lin CW, Tseng SW, Yang SF, et al. Role of lipocalin 2 and
its complex with matrix metalloproteinase-9 in oral can-
cer. Oral Dis. 2012;18:734740.
10. Campestre C, Agamennone M, Tauro M, et al.
Phosphonate emerging zinc binding group in matrix
metalloproteinase inhibitors. Curr Drug Targets.
2015;16:16341644.
11. Fields GB. New strategies for targeting matrix metallo-
proteinases. Matrix Biol. 2015;4446:239246.
This article gives an overview of new strategies for
targeting MMPs.
12. Li NG, Tang YP, Duan JA, et al. Matrix metalloproteinase
inhibitors: a patent review (2011 - 2013). Expert Opin
Ther Pat. 2014;24:10391052.
13. Cao J, Sato H, Takino T, et al. The C-terminal region of
membrane type matrix metalloproteinase is a functional
transmembrane domain required for pro-gelatinase A
activation. J Biol Chem. 1995;270:801805.
14. Ra HJ, Parks WC. Control of matrix metalloproteinase
catalytic activity. Matrix Biol. 2007;26:587596.
15. Fanjul-Fernandez M, Folgueras AR, Cabrera S, et al. Matrix
metalloproteinases: evolution, gene regulation and func-
tional analysis in mouse models. Biochim Biophys Acta.
2010;1803:319.
EXPERT OPINION ON DRUG METABO LISM & TOXICOLOGY 197
Downloaded by [Institute of Software] at 00:58 15 February 2016
16. Barksby HE, Milner JM, Patterson AM, et al. Matrix metal-
loproteinase 10 promotion of collagenolysis via procolla-
genase activation: implications for cartilage degradation
in arthritis. Arthritis Rheum. 2006;54:32443253.
17. Geurts N, Martens E, Van Aelst I, et al. Beta-hematin
interaction with the hemopexin domain of gelatinase
B/MMP-9 provokes autocatalytic processing of the pro-
peptide, thereby priming activation by MMP-3.
Biochemistry. 2008;47:26892699.
18. Uria JA, Lopez-Otin C. Matrilysin-2, a new matrix metal-
loproteinase expressed in human tumors and showing
the minimal domain organization required for secretion,
latency, and activity. Cancer Res. 2000;60:47454751.
19. Gomis-Ruth FX. Catalytic domain architecture of metzin-
cin metalloproteases. J Biol Chem. 2009;284:15353
15357.
20. Zucker S, Pei D, Cao J, et al. Membrane type-matrix
metalloproteinases (MT-MMP). Curr Top Dev Biol.
2003;54:174.
21. Rohani MG, Parks WC. Matrix remodeling by MMPs dur-
ing wound repair. Matrix Biol. 2015;4446:113121.
22. Nagase H, Woessner JF Jr. Matrix metalloproteinases. J
Biol Chem. 1999;274:2149121494.
23. Page-McCaw A, Ewald AJ, Werb Z. Matrix metalloprotei-
nases and the regulation of tissue remodelling. Nat Rev
Mol Cell Biol. 2007;8:221233.
24. Kleiner DE, Stetler-Stevenson WG. Matrix metalloprotei-
nases and metastasis. Cancer Chemother Pharmacol.
1999;43:S4251.
•• This article gives an overview of the mechanisms of
cancer metastasis.
25. Deryugina EI, Quigley JP. Tumor angiogenesis: MMP-
mediated induction of intravasation- and metastasis-sus-
taining neovasculature. Matrix Biol. 2015;4446:94112.
26. Shay G, Lynch CC, Fingleton B. Moving targets: emerging
roles for MMPs in cancer progression and metastasis.
Matrix Biol. 2015;4446:200206.
27. Loffek S, Schilling O, Franzke CW. Series matrix metallo-
proteinases in lung health and disease: biological role of
matrix metalloproteinases: a critical balance. Eur Respir J.
2011;38:191208.
28. Murphy G. Tissue inhibitors of metalloproteinases.
Genome Biol. 2011;12:233.
29. Tchetverikov I, Verzijl N, Huizinga TW, et al. Active MMPs
captured by alpha 2 macroglobulin as a marker of dis-
ease activity in rheumatoid arthritis. Clin Exp Rheumatol.
2003;21:711718.
30. Remillard TC, Bratslavsky G, Jensen-Taubman S, et al.
Molecular mechanisms of tissue inhibitor of metallopro-
teinase 2 in the tumor microenvironment. Mol Cell Ther.
2014;2:17.
This article gives an overview of the mechanisms of
TIMP-2 in cancer.
31. Henriet P, Blavier L, Declerck YA. Tissue inhibitors of
metalloproteinases (TIMP) in invasion and proliferation.
APMIS. 1999;107:111119.
32. Bode W, Fernandez-Catalan C, Nagase H, et al.
Endoproteinase-protein inhibitor interactions. APMIS.
1999;107:310.
33. Itoh Y. Metalloproteinase binding proteins:
WO2009097397. Expert Opin Ther Pat. 2010;20:10911095.
34. Palei AC, Granger JP, Tanus-Santos JE. Matrix metallopro-
teinases as drug targets in preeclampsia. Curr Drug
Targets. 2013;14:325334.
35. Baker AH, Edwards DR, Murphy G. Metalloproteinase
inhibitors: biological actions and therapeutic opportu-
nities. J Cell Sci. 2002;115:37193727.
36. Valente P, Fassina G, Melchiori A, et al. TIMP-2 over-
expression reduces invasion and angiogenesis and pro-
tects B16F10 melanoma cells from apoptosis. Int J
Cancer. 1998;75:246253.
37. Bond M, Murphy G, Bennett MR, et al. Tissue inhibitor of
metalloproteinase-3 induces a Fas-associated death
domain-dependent type II apoptotic pathway. J Biol
Chem. 2002;277:1378713795.
38. Ahonen M, Poukkula M, Baker AH, et al. Tissue inhibitor
of metalloproteinases-3 induces apoptosis in melanoma
cells by stabilization of death receptors. Oncogene.
2003;22:21212134.
39. Nelson AR, Fingleton B, Rothenberg ML, et al. Matrix
metalloproteinases: biologic activity and clinical implica-
tions. J Clin Oncol. 2000;18:11351149.
40. Betz M, Huxley P, Davies SJ, et al. 1.8-A crystal structure
of the catalytic domain of human neutrophil collagenase
(matrix metalloproteinase-8) complexed with a peptido-
mimetic hydroxamate primed-side inhibitor with a dis-
tinct selectivity profile. Eur J Biochem. 1997;247:356363.
41. Macaulay VM, OByrne KJ, Saunders MP, et al. Phase I
study of intrapleural batimastat (BB-94), a matrix metal-
loproteinase inhibitor, in the treatment of malignant
pleural effusions. Clin Cancer Res. 1999;5:513520.
42. Acharya MR, Venitz J, Figg WD, et al. Chemically modified
tetracyclines as inhibitors of matrix metalloproteinases.
Drug Resist Updat. 2004;7:195208.
43. Brown PD. Clinical studies with matrix metalloproteinase
inhibitors. APMIS. 1999;107:174180.
44. Davies B, Brown PD, East N, et al. A synthetic matrix
metalloproteinase inhibitor decreases tumor burden
and prolongs survival of mice bearing human ovarian
carcinoma xenografts. Cancer Res. 1993;53:20872091.
45. Chirivi RG, Garofalo A, Crimmin MJ, et al. Inhibition of the
metastatic spread and growth of B16-BL6 murine mela-
noma by a synthetic matrix metalloproteinase inhibitor.
Int J Cancer. 1994;58:460464.
46. Sledge GW Jr, Qulali M, Goulet R, et al. Effect of matrix
metalloproteinase inhibitor batimastat on breast cancer
regrowth and metastasis in athymic mice. J Natl Cancer
Inst. 1995;87:15461550.
47. Watson SA, Morris TM, Robinson G, et al. Inhibition of
organ invasion by the matrix metalloproteinase inhibitor
batimastat (BB-94) in two human colon carcinoma
metastasis models. Cancer Res. 1995;55:36293633.
48. Eccles SA, Box GM, Court WJ, et al. Control of lymphatic
and hematogenous metastasis of a rat mammary carci-
noma by the matrix metalloproteinase inhibitor batima-
stat (BB-94). Cancer Res. 1996;56:28152822.
49. Low JA, Johnson MD, Bone EA, et al. The matrix metallo-
proteinase inhibitor batimastat (BB-94) retards human
breast cancer solid tumor growth but not ascites forma-
tion in nude mice. Clin Cancer Res. 1996;2:12071214.
50. Zervos EE, Norman JG, Gower WR, et al. Matrix metallo-
proteinase inhibition attenuates human pancreatic
198 J.-S. YANG ET AL.
Downloaded by [Institute of Software] at 00:58 15 February 2016
cancer growth in vitro and decreases mortality and
tumorigenesis in vivo. J Surg Res. 1997;69:367371.
51. Bu W, Tang ZY, Sun FX, et al. Effects of matrix metallo-
proteinase inhibitor BB-94 on liver cancer growth and
metastasis in a patient-like orthotopic model LCI-D20.
Hepatogastroenterology. 1998;45:10561061.
52. Wylie S, MacDonald IC, Varghese HJ, et al. The matrix
metalloproteinase inhibitor batimastat inhibits angiogen-
esis in liver metastases of B16F1 melanoma cells. Clin Exp
Metastasis. 1999;17:111117.
53. Wada N, Otani Y, Kubota T, et al. Reduced angiogenesis
in peritoneal dissemination of gastric cancer through
gelatinase inhibition. Clin Exp Metastasis. 2003;20:431
435.
54. Wojtowicz-Praga S, Low J, Marshall J, et al. Phase I trial of
a novel matrix metalloproteinase inhibitor batimastat
(BB-94) in patients with advanced cancer. Invest New
Drugs. 1996;14:193202.
55. Beattie GJ, Smyth JF. Phase I study of intraperitoneal
metalloproteinase inhibitor BB94 in patients with malig-
nant ascites. Clin Cancer Res. 1998;4:18991902.
56. Wojtowicz-Praga SM, Dickson RB, Hawkins MJ. Matrix
metalloproteinase inhibitors. Invest New Drugs.
1997;15:6175.
57. Coussens LM, Fingleton B, Matrisian LM. Matrix metallo-
proteinase inhibitors and cancer: trials and tribulations.
Science. 2002;295:23872392.
58. Wall L, Talbot DC, Bradbury P, et al. A phase I and
pharmacological study of the matrix metalloproteinase
inhibitor BB-3644 in patients with solid tumours. Br J
Cancer. 2004;90:800804.
59. Santos O, McDermott CD, Daniels RG, et al. Rodent phar-
macokinetic and anti-tumor efficacy studies with a series
of synthetic inhibitors of matrix metalloproteinases. Clin
Exp Metastasis. 1997;15:499508.
60. Shalinsky DR, Brekken J, Zou H, et al. Antitumor efficacy
of AG3340 associated with maintenance of minimum
effective plasma concentrations and not total daily
dose, exposure or peak plasma concentrations. Invest
New Drugs. 1998;16:303313.
61. Shalinsky DR, Brekken J, Zou H, et al. Broad antitumor
and antiangiogenic activities of AG3340, a potent and
selective MMP inhibitor undergoing advanced oncology
clinical trials. Ann N Y Acad Sci. 1999;878:236270.
62. Shalinsky DR, Brekken J, Zou H, et al. Marked antiangio-
genic and antitumor efficacy of AG3340 in chemoresis-
tant human non-small cell lung cancer tumors: single
agent and combination chemotherapy studies. Clin
Cancer Res. 1999;5:19051917.
63. Price A, Shi Q, Morris D, et al. Marked inhibition of tumor
growth in a malignant glioma tumor model by a novel
synthetic matrix metalloproteinase inhibitor AG3340. Clin
Cancer Res. 1999;5:845854.
64. Hidalgo M, Eckhardt SG. Development of matrix metallo-
proteinase inhibitors in cancer therapy. J Natl Cancer
Inst. 2001;93:178193.
65. Hande KR, Collier M, Paradiso L, et al. Phase I and phar-
macokinetic study of prinomastat, a matrix metallopro-
tease inhibitor. Clin Cancer Res. 2004;10:909915.
66. Bissett D, OByrne KJ, von Pawel J, et al. Phase III study of
matrix metalloproteinase inhibitor prinomastat in non-
small-cell lung cancer. J Clin Oncol. 2005;23:842849.
67. Rowinsky EK, Humphrey R, Hammond LA, et al. Phase I
and pharmacologic study of the specific matrix metallo-
proteinase inhibitor BAY 12-9566 on a protracted oral
daily dosing schedule in patients with solid malignan-
cies. J Clin Oncol. 2000;18:178186.
68. Gatto C, Rieppi M, Borsotti P, et al. BAY 12-9566, a novel
inhibitor of matrix metalloproteinases with antiangio-
genic activity. Clin Cancer Res. 1999;5:36033607.
69. Molina JR, Reid JM, Erlichman C, et al. A phase I and
pharmacokinetic study of the selective, non-peptidic
inhibitor of matrix metalloproteinase BAY 12-9566 in
combination with etoposide and carboplatin.
Anticancer Drugs. 2005;16:9971002.
70. Heath EI, OReilly S, Humphrey R, et al. Phase I trial of the
matrix metalloproteinase inhibitor BAY12-9566 in
patients with advanced solid tumors. Cancer
Chemother Pharmacol. 2001;48:269274.
71. Hirte H, Vergote IB, Jeffrey JR, et al. A phase III rando-
mized trial of BAY 12-9566 (tanomastat) as maintenance
therapy in patients with advanced ovarian cancer
responsive to primary surgery and paclitaxel/platinum
containing chemotherapy: a National Cancer Institute of
Canada Clinical Trials Group Study. Gynecol Oncol.
2006;102:300308.
72. Ferrante K, Winograd B, Canetta R. Promising new devel-
opments in cancer chemotherapy. Cancer Chemother
Pharmacol. 1999;43:S6168.
73. Rizvi NA, Humphrey JS, Ness EA, et al. A phase I study of
oral BMS-275291, a novel nonhydroxamate sheddase-
sparing matrix metalloproteinase inhibitor, in patients
with advanced or metastatic cancer. Clin Cancer Res.
2004;10:19631970.
74. Miller KD, Saphner TJ, Waterhouse DM, et al. A rando-
mized phase II feasibility trial of BMS-275291 in patients
with early stage breast cancer. Clin Cancer Res.
2004;10:19711975.
75. Leighl NB, Paz-Ares L, Douillard JY, et al. Randomized
phase III study of matrix metalloproteinase inhibitor
BMS-275291 in combination with paclitaxel and carbo-
platin in advanced non-small-cell lung cancer: National
Cancer Institute of Canada-Clinical Trials Group Study
BR.18. J Clin Oncol. 2005;23:28312839.
76. Levitt NC, Eskens FA, OByrne KJ, et al. Phase I and phar-
macological study of the oral matrix metalloproteinase
inhibitor, MMI270 (CGS27023A), in patients with advanced
solid cancer. Clin Cancer Res. 2001;7:19121922.
77. Eatock M, Cassidy J, Johnson J, et al. A dose-finding and
pharmacokinetic study of the matrix metalloproteinase
inhibitor MMI270 (previously termed CGS27023A) with 5-
FU and folinic acid. Cancer Chemother Pharmacol.
2005;55:3946.
78. Rao BG. Recent developments in the design of specific
matrix metalloproteinase inhibitors aided by structural
and computational studies. Curr Pharm Des.
2005;11:295322.
79. Golub LM, Lee HM, Ryan ME, et al. Tetracyclines inhibit
connective tissue breakdown by multiple non-antimicro-
bial mechanisms. Adv Dent Res. 1998;12:1226.
80. Golub LM, Ramamurthy N, McNamara TF, et al.
Tetracyclines inhibit tissue collagenase activity. A new
mechanism in the treatment of periodontal disease. J
Periodontal Res. 1984;19:651655.
EXPERT OPINION ON DRUG METABO LISM & TOXICOLOGY 199
Downloaded by [Institute of Software] at 00:58 15 February 2016
81. Fife RS, Rougraff BT, Proctor C, et al. Inhibition of prolif-
eration and induction of apoptosis by doxycycline in
cultured human osteosarcoma cells. J Lab Clin Med.
1997;130:530534.
82. Gilbertson-Beadling S, Powers EA, Stamp-Cole M, et al.
The tetracycline analogs minocycline and doxycycline
inhibit angiogenesis in vitro by a non-metalloprotei-
nase-dependent mechanism. Cancer Chemother
Pharmacol. 1995;36:418424.
83. Fife RS, Sledge GW Jr. Effects of doxycycline on in vitro
growth, migration, and gelatinase activity of breast car-
cinoma cells. J Lab Clin Med. 1995;125:407411.
84. Duivenvoorden WC, Hirte HW, Singh G. Transforming
growth factor beta1 acts as an inducer of matrix metal-
loproteinase expression and activity in human bone-
metastasizing cancer cells. Clin Exp Metastasis.
1999;17:2734.
85. Lokeshwar BL. MMP inhibition in prostate cancer. Ann N
Y Acad Sci. 1999;878:271289.
86. Fife RS, Sledge GW Jr, Roth BJ, et al. Effects of doxycy-
cline on human prostate cancer cells in vitro. Cancer Lett.
1998;127:3741.
87. Duivenvoorden WC, Hirte HW, Singh G. Use of tetracy-
cline as an inhibitor of matrix metalloproteinase activity
secreted by human bone-metastasizing cancer cells.
Invasion Metastasis. 1997;17:312322.
88. Huie M, Oettel K, Van Ummersen L, et al. Phase II study of
interferon-alpha and doxycycline for advanced renal cell
carcinoma. Invest New Drugs. 2006;24:255260.
89. Lokeshwar BL, Selzer MG, Zhu BQ, et al. Inhibition of cell
proliferation, invasion, tumor growth and metastasis by
an oral non-antimicrobial tetracycline analog (COL-3) in a
metastatic prostate cancer model. Int J Cancer.
2002;98:297309.
90. Syed S, Takimoto C, Hidalgo M, et al. A phase I and
pharmacokinetic study of Col-3 (Metastat), an oral tetra-
cycline derivative with potent matrix metalloproteinase
and antitumor properties. Clin Cancer Res. 2004;10:6512
6521.
91. Chu QS, Forouzesh B, Syed S, et al. A phase II and
pharmacological study of the matrix metalloproteinase
inhibitor (MMPI) COL-3 in patients with advanced soft
tissue sarcomas. Invest New Drugs. 2007;25:359367.
92. Farina AR, Tacconelli A, Teti A, et al. Tissue inhibitor of
metalloproteinase-2 protection of matrix metalloprotei-
nase-2 from degradation by plasmin is reversed by diva-
lent cation chelator EDTA and the bisphosphonate
alendronate. Cancer Res. 1998;58:29572960.
93. Teronen O, Heikkila P, Konttinen YT, et al. MMP inhibition
and downregulation by bisphosphonates. Ann N Y Acad
Sci. 1999;878:453465.
94. Dedes PG, Gialeli C, Tsonis AI, et al. Expression of matrix
macromolecules and functional properties of breast can-
cer cells are modulated by the bisphosphonate zoledro-
nic acid. Biochim Biophys Acta. 2012;1820:19261939.
95. Lipton A. Bones, breasts, and bisphosphonates: rationale
for the use of zoledronic acid in advanced and early
breast cancer. Breast Cancer (Dove Med Press).
2011;3:17.
96. Rosenthal EL, Matrisian LM. Matrix metalloproteases in
head and neck cancer. Head Neck. 2006;28:639648.
97. Dufour A, Zucker S, Sampson NS, et al. Role of matrix
metalloproteinase-9 dimers in cell migration: design of
inhibitory peptides. J Biol Chem. 2010;285:3594435956.
98. Sela-Passwell N, Rosenblum G, Shoham T, et al. Structural
and functional bases for allosteric control of MMP activ-
ities: can it pave the path for selective inhibition?
Biochim Biophys Acta. 2010;1803:2938.
200 J.-S. YANG ET AL.
Downloaded by [Institute of Software] at 00:58 15 February 2016
... Not Only through Its Known MOAs. COL-3 is a known MMP inhibitor, with inhibitory effects against multiple MMP molecules including MMP-1, MMP-2, MMP-3, MMP-7, MMP-9, and MMP-12 (24,25). To test whether the higher efficacy of COL-3 in R1-D567 vs. R1-AD1 cells was due to differences in the activities of MMPs, we analyzed microarray gene expression data to assess mRNA levels of 22 MMP genes (Fig. 4). ...
Article
Full-text available
Prostate cancer (PC) is the most frequently diagnosed malignancy and a leading cause of cancer deaths in US men. Many PC cases metastasize and develop resistance to systemic hormonal therapy, a stage known as castration-resistant prostate cancer (CRPC). Therefore, there is an urgent need to develop effective therapeutic strategies for CRPC. Traditional drug discovery pipelines require significant time and capital input, which highlights a need for novel methods to evaluate the repositioning potential of existing drugs. Here, we present a computational framework to predict drug sensitivities of clinical CRPC tumors to various existing compounds and identify treatment options with high potential for clinical impact. We applied this method to a CRPC patient cohort and nominated drugs to combat resistance to hormonal therapies including abiraterone and enzalutamide. The utility of this method was demonstrated by nomination of multiple drugs that are currently undergoing clinical trials for CRPC. Additionally, this method identified the tetracycline derivative COL-3, for which we validated higher efficacy in an isogenic cell line model of enzalutamide-resistant vs. enzalutamide-sensitive CRPC. In enzalutamide-resistant CRPC cells, COL-3 displayed higher activity for inhibiting cell growth and migration, and for inducing G1-phase cell cycle arrest and apoptosis. Collectively, these findings demonstrate the utility of a computational framework for independent validation of drugs being tested in CRPC clinical trials, and for nominating drugs with enhanced biological activity in models of enzalutamide-resistant CRPC. The efficiency of this method relative to traditional drug development approaches indicates a high potential for accelerating drug development for CRPC.
... The tissue inhibitor of metalloproteinases (TIMPs), including TIMP1, 2, 3, and 4, are endogenous regulators of matrix metalloproteinases (MMPs) [5][6][7][8]. TIMPs have been implicated in extracellular matrix degradation, tissue remodeling, cancer cell invasion, and metastasis, and an imbalance between TIMPs and MMPs activity may have implications for cancer progression [9][10][11]. TIMP3 is unique among the mammalian TIMPs, differing from other TIMPs in that it is tightly bound to the extracellular matrix and it has a broader inhibitory activity against MMPs and inhibits it's closely related a disintegrin and metalloproteinases [12]. ...
Article
Full-text available
The tissue inhibitors of metalloproteinases-3 (TIMP3) are not only endogenous regulators of matrix metalloproteinases (MMPs), but also induce apoptosis and inhibit endothelial cell migration and angiogenesis. The focus of this study was to investigate the relationship between TIMP3 genetic polymorphisms and biochemical recurrence and clinicopathological features of prostate cancer. The TIMP3 rs9619311, rs9862, and rs11547635 genetic polymorphisms were analyzed by real-time polymerase chain reaction to determine their genotypic distributions in 579 patients with prostate cancer. This study found that individuals with the TIMP3 rs9619311 TC or TC + CC genotypes have a significantly higher risk of biochemical recurrence of prostate cancer (p = 0.036 and 0.033, respectively). Moreover, in the multivariate analysis, our results showed that pathologic Gleason grade, pathologic T stage, seminal vesicle invasion, lymphovascular invasion, and TIMP3 rs9619311 were associated with increased odds of biochemical recurrence. Patients with a PSA concentration under 7 ng/mL that were found to have the TIMP3 rs9619311 genetic polymorphism were associated with Gleason total score upgrade (p = 0.012) and grade group upgrade (p = 0.023). Compared with the CC homozygous, the TIMP3 rs9862 CT + TT polymorphic variant was found to be associated with clinically advanced tumor stage (p = 0.030) and Gleason total score upgrade (p = 0.002) in prostate cancer patients. In conclusion, the results of our study demonstrated that the TIMP3 rs9619311 genetic polymorphism was significantly associated with susceptibility to biochemical recurrence of prostate cancer. TIMP3 genetic polymorphisms, especially rs9619311, can serve as key predictors of biochemical recurrence and disease prognosis of prostate cancer.
... Matrix metalloproteinases, a family of zinc-dependent proteinase, have long been associated with tumor cell invasion and metastasis for its ability in activating cancer cells, modulating immune function, degrading extracellular matrix and basement membrane. [25][26][27][28] In particular, matrix metalloproteinase-9 (MMP-9) has been widely investigated to relate to the pathology of cancer. 29-31 TPA (12-O-Tetradecanoylphorbol-13-acetate), a phorbol ester, has been well-documented on cancer metastasis in several cancer types. ...
Article
Diphenyl difluoroketone (EF‐24), a synthetic curcumin analog, has enhanced bioavailability over curcumin. EF‐24 acts more powerful bioactivity for anti‐inflammatory and anti‐cancer activity. However, the effects and mechanism of EF‐24 on cervical cancer has not been fully investigated. Herein, this study evaluated the effects of EF‐24 on TPA‐induced cellular migration of cervical cancer. The results showed that EF‐24 substantially reduced the cellular migration and cellular invasion of the HeLa and SiHa cells. Moreover, gelatin zymography, western blotting analyses and real‐time PCR revealed that EF‐24 suppressed Matrix metalloproteinase‐9 (MMP‐9) activity, protein expression and mRNA levels. Mechanistically, EF‐24 inhibited the phosphorylation of the p38 signaling pathway. In conclusion, EF‐24 inhibited TPA‐induced cellular migration and cellular invasion of cervical cancer cell lines through modulating MMP‐9 expression via downregulating signaling p38 pathway and EF‐24 may have potential to serve as a chemopreventive agent of cervical cancer.
... The matrix metalloproteinases (MMPs) are a zinc-dependent endopeptidases family which is involved in extracellular matrix (ECM) degradation and tissue remodeling [18][19][20][21]. MMP-11, or stromelysin 3, is a proteolytic enzyme which belongs to the MMP family [22][23][24]. ...
Article
Full-text available
Colorectal cancer (CRC) is the third most common cause of cancer mortality worldwide and the most prevalent cancer in Taiwan. The matrix metalloproteinase (MMP)-11 is a proteolytic enzyme of the MMP family which is involved in extracellular matrix degradation and tissue remodeling. In this study, we focused on the associations of MMP-11 single-nucleotide polymorphisms (SNPs) with CRC susceptibility and clinicopathological characteristics. The MMP-11 SNPs rs131451, rs738791, rs2267029, rs738792, and rs28382575 in 479 controls and 479 patients with CRC were analyzed with real-time polymerase chain reaction. We found that the MMP-11 SNP rs738792 “TC + CC” genotype was significantly associated with perineural invasion in colon cancer patients after controlling for clinical parameters [OR (95% CI) = 1.783 (1.074–2.960); p = 0.025]. The MMP-11 rs131451 “TC + CC” genotypic variants were correlated with greater tumor T status [OR (95% CI):1.254 (1.025–1.534); p = 0.028] and perineural invasion [OR (95% CI):1.773 (1.027–3.062); p = 0.040) in male CRC patients. Furthermore, analyses of The Cancer Genome Atlas (TCGA) revealed that MMP-11 levels were upregulated in colorectal carcinoma tissue compared with normal tissues and were correlated with advanced stage, larger tumor sizes, and lymph node metastasis. Moreover, the data from the Genotype-Tissue Expression (GTEx) database exhibited that the MMP-11 rs738792 “CC” and “CT” genotypic variants have higher MMP-11 expression than the “TT” genotype. In conclusion, our results have demonstrated that the MMP-11 SNPs rs738792 and rs131451 may have potential to provide biomarkers to evaluate CRC disease progression, and the MMP-11 rs131451 polymorphism may shed light on sex discrepancy in CRC development and prognosis.
... MMPs are involved in cell growth, invasion, survival, and adhesion in biological and pathological conditions, and then their activation Ivyspring International Publisher can cause cell proliferation, invasion and metastasis of cancer cells [4][5][6]. Tissue inhibitors of MMPs (TIMPs) are endogenous inhibitors of MMPs, and imbalance between the activities of MMPs and TIMPs may have an impact on cancer progression [7][8][9][10]. The TIMPs are known as a family of at least four 20 to 29 kDa proteins (TIMPs 1-4) which reversibly inhibit the MMPs [11]. ...
Article
Full-text available
Single nucleotide polymorphisms (SNPs) of tissue inhibitor of metalloproteinases-3 (TIMP-3) have been revealed to be related to various cancers. To date, no study explores the relationships between TIMP-3 polymorphisms and uterine cervical cancer. The purposes of this research were to investigate the associations among genetic variants of TIMP-3 and development and clinicopathological factors of uterine cervical cancer, and patient 5 years survival in Taiwanese women. The study included 123 patients with invasive cancer and 97 with precancerous lesions of uterine cervix, and 300 control women. TIMP-3 polymorphisms rs9619311, rs9862 and rs11547635 were checked and their genotypic distributions were determined by real-time polymerase chain reaction. It showed that women with genotypes CT/TT in rs9862 were found to display a higher risk of developing cervical cancer with moderate and poor cell differentiation. Moreover, it revealed that cervical cancer patients carrying genotypes CC in rs9619311 exhibited a poorer 5 years survival, as compared to those with TT/TC in Taiwanese women, using univariate analysis. In addition, pelvic lymph node metastasis was determined to independently predict 5 years survival in cervical cancer patients using multivariate analysis. Conclusively, TIMP-3 SNPs polymorphisms rs9619311 are related to cervical patient survival in Taiwanese women.
... MMPs play an important role in cancer progression, and different strategies have been developed to inhibit their expression and enzymatic activity including synthetic MMP inhibitors (sMMPIs). sMMPIs can be grouped into peptidomimetic, nonpeptidomimetic, chemically modified tetracyclines, and thiiranebased slow inhibitors (Li et al., 2013;Yang et al., 2016;Marusak et al., 2016;Meisel and Chang, 2017). Some examples of the former are displayed in Table 4. ...
Article
Full-text available
Cancer is still one of the leading causes of death worldwide. This great mortality is due to its late diagnosis when the disease is already at advanced stages. Although the efforts made to develop more effective treatments, around 90% of cancer deaths are due to metastasis that confers a systemic character to the disease. Likewise, matrix metalloproteinases (MMPs) are endopeptidases that participate in all the events of the metastatic process. MMPs’ augmented concentrations and an increased enzymatic activity have been considered bad prognosis markers of the disease. Therefore, synthetic inhibitors have been created to block MMPs’ enzymatic activity. However, they have been ineffective in addition to causing considerable side effects. On the other hand, nanotechnology offers the opportunity to formulate therapeutic agents that can act directly on a target cell, avoiding side effects and improving the diagnosis, follow-up, and treatment of cancer. The goal of the present review is to discuss novel nanotechnological strategies in which MMPs are used with theranostic purposes and as therapeutic targets to control cancer progression.
Article
Full-text available
The tumor microenvironment (TME) is characterized by an acidic pH and low oxygen concentrations. Hypoxia induces neoplastic cell evasion of the immune surveillance, rapid DNA repair, metabolic reprogramming, and metastasis, mainly as a response to the hypoxic inducible factors (HIFs). Likewise, cancer cells increase matrix metalloproteinases’ (MMPs) expression in response to TME conditions, allowing them to migrate from the primary tumor to different tissues. Since HIFs and MMPs are augmented in the hypoxic TME, it is easy to consider that HIFs participate directly in their expression regulation. However, not all MMPs have a hypoxia response element (HRE)-HIF binding site. Moreover, different transcription factors and signaling pathways activated in hypoxia conditions through HIFs or in a HIF-independent manner participate in MMPs’ transcription. The present review focuses on MMPs’ expression in normal and hypoxic conditions, considering HIFs and a HIF-independent transcription control. In addition, since the hypoxic TME causes resistance to anticancer conventional therapy, treatment approaches using MMPs as a target alone, or in combination with other therapies, are also discussed.
Article
Matrix metalloproteinases (MMPs) are proteolytic enzymes that aid in extracellular matrix (ECM) remodeling. MMPs destroy the extracellular matrix, causing tumor growth and metastasis. MMPs are involved in the spread and metastasis of oral cancer. High levels of MMPs and oral squamous cell carcinoma have been linked to cancer prognosis. Modern medicine aims to prevent the illness from spreading through early intervention and examining changes in MMP genes. MMP gene polymorphism has recently been identified as one of the factors predicting susceptibility or risk in the development of oral carcinoma. This review aims to provide insight into the function of MMP subtypes involved in cancer. The genetic polymorphism in MMP genes and its predictive value in risk evaluation have been elaborated. Novel personalized therapeutic approaches for oral cancer, like the use of MMP inhibitors, nanoparticle-mediated targeting of MMP, or gene silencing by microRNA, can be designed.
Article
The pro- or antitumoral properties of nitric oxide (NO) are dependent on local concentration, redox state, cellular status, duration of exposure and compartmentalization of NO generation. The intricate network of the tumor microenvironment (TME) is constituted by tumor cells, stromal and immune cells surrounded by active components of extracellular matrix that influence the biological behavior and, consequently, the treatment and prognosis of cancer. The review describes critical events in the crosstalk of cellular and stromal components in the TME, with special emphasis in the impact of NO generation in the regulation of hepatocarcinoma (HCC). The increased expression of nitric oxide synthase (NOS) in tumors and NO-end products in plasma has been associated with poor prognosis of cancer. We have assessed the level of the different isoforms of NOS in tumors and its relation to cell proliferation and death markers, and cell death receptor expression in tumors, and apoptotic markers and ligands of TNF-α receptor family in blood from a cohort of patients with HCC from different etiologies submitted to orthotopic liver transplantation (OLT). The high levels of NOS2 in tumors were associated with low plasma concentration of apoptotic markers (M30 and M65), FasL and TNF-α in HCV patients. By contrast, the low levels of NOS2 in tumors from alcohol-derived patients was associated with increased Trail-R1 expression in tumors, and circulating Trail levels compared to observed in plasma from HCV- and alcohol + HCV-derived patients. This study reinforces the association between increased NOS2 expression and potential risk of low patients’ survival in HCC. However, a differential functional relevance of NOS expression in HCC seems to be influenced by etiologies.
Article
Objective: To evaluate the associations between dipeptidyl peptidase IV (DPP4) single nucleotide polymorphism (SNP) and clinicopathological characters of oral cancer. Methods: Four loci of DPP4 SNPs (rs7608798 A/G, rs3788979 C/T, rs2268889 T/C, and rs6741949 G/C) were genotyped by using the TaqMan allelic discrimination in 1238 oral cancers patients and 1197 non-cancer individuals. Results: The percentage of DPP4 SNP rs2268889 TC+CC was significantly higher in the oral cancer participants compared to the control group (odds ratio (OR): 1.178, 95% confidence interval (CI): 1.004-1.382, P = 0.045). Among 1676 smokers, DPP4 polymorphisms carriers with betel quid chewing were found to have a 8.785- to 10.903-fold risk to have oral cancer compared to DPP4 wild-type carriers without betel quid chewing. Similar trend was found in individuals with alcohol consumption. Moreover, the oral cancer individuals without cigarette smoking history with at least 1 varied C allele of DPP4 rs2268889 had a significantly higher percentage of large tumor size with the wild-type TT homozygote (P= 0.011). Conclusions: The DPP4 SNP may correlate to the development of oral cancer in those with cigarette smoking and alcohol consumption. Besides, the DPP4 SNP rs2268889 could relate to worse clinical course of oral cancer in non-smokers.
Article
Full-text available
Metastasis is a distinct stage of cancer progression that requires the development of angiogenic blood vessels serving as conduits for tumor cell dissemination. An accumulated body of evidence indicates that metastasis-supporting neovasculature should possess certain structural characteristics allowing for the process of tumor cell intravasation, an active entry of cancer cells into the vessel interior. It appears that the development of tumor vessels with lumens of a distinctive size and their structure supported by a discontinuous pericyte coverage, together constitute critical microarchitectural requirements to: (a) provide accessible points for vessel wall penetration by primary tumor cells; (b) provide enough lumen space for a tumor cell or cell aggregate upon intravasation; and (c) allow for sufficient rate of blood flow to carry away intravasated cells from the primary tumor to the next, proximal or distal site. This review will primarily focus on the functional roles of matrix metalloproteinases (MMPs), which catalytically trigger the development of an intravasation-sustaining neovasculature at the early stages of tumor growth and are also required for the maintenance of a metastasis-supporting state of blood vessels at later stages of cancer progression. Copyright © 2015. Published by Elsevier B.V.
Article
Full-text available
There has been a recent paradigm shift in the way we target cancer, drawing a greater focus on the role of the tumor microenvironment (TME) in cancer development, progression and metastasis. Within the TME, there is a crosstalk in signaling and communication between the malignant cells and the surrounding extracellular matrix. Matrix metalloproteinases (MMPs) are zinc-dependent endoproteases that have the ability to degrade the matrix surrounding a tumor and mediate tumor growth, angiogenesis and metastatic disease. Their endogenous inhibitors, the Tissue Inhibitors of Metalloproteinases (TIMPs), primarily function to prevent degradation of the ECM via inhibition of MMPs. However, recent studies demonstrate that TIMP family members also possess MMP-independent functions. One TIMP member in particular, TIMP-2, has many distinct properties and functions, that occur independent of MMP inhibition, including the inhibition of tumor growth and reduction of angiogenesis through decreased endothelial cell proliferation and migration. The MMP-independent molecular mechanisms and signaling pathways elicited by TIMP-2 in the TME are described in this review.
Article
Full-text available
Repair following injury involves a range of processes - such as re-epithelialization, scar formation, angiogenesis, inflammation, and more - that function, often together, to restore tissue architecture. MMPs carry out diverse roles in all of these activities. In this article, we discuss how specific MMPs act on ECM during two critical repair processes: re-epithelialization and resolution of scar tissue. For wound closure, we discuss how two MMPs - MMP1 in human epidermis and MMP7 in mucosal epithelia - facilitate re-epithelialization by cleaving different ECM or ECM-associated proteins to affect similar integrin:matrix adhesion. In scars and fibrotic tissues, we discuss that a variety of MMPs carry out a diverse range of activities that can either promote or limit ECM deposition. However, few of these MMP-driven activities have been demonstrated to be due a direct action on ECM. Copyright © 2015. Published by Elsevier B.V.
Article
: Tumor progression is a complex, multistage process by which a normal cell undergoes genetic changes that result in phenotypic alterations and the acquisition of the ability to spread and colonize distant sites in the body. Although many factors regulate malignant tumor growth and spread, interactions between a tumor and its surrounding microenvironment result in the production of important protein products that are crucial to each step of tumor progression. The matrix metalloproteinases (MMPs) are a family of degradative enzymes with clear links to malignancy. These enzymes are associated with tumor cell invasion of the basement membrane and stroma, blood vessel penetration, and metastasis. They have more recently been implicated in primary and metastatic tumor growth and angiogenesis, and they may even have a role in tumor promotion. This review outlines our current understanding of the MMP family, including the association of particular MMPs with malignant phenotypes and the role of MMPs in sp...
Article
Each year the American Cancer Society estimates the numbers of new cancer cases and deaths that will occur in the United States in the current year and compiles the most recent data on cancer incidence, mortality, and survival. Incidence data were collected by the National Cancer Institute (Surveillance, Epidemiology, and End Results [SEER] Program), the Centers for Disease Control and Prevention (National Program of Cancer Registries), and the North American Association of Central Cancer Registries. Mortality data were collected by the National Center for Health Statistics. A total of 1,658,370 new cancer cases and 589,430 cancer deaths are projected to occur in the United States in 2015. During the most recent 5 years for which there are data (2007-2011), delay-adjusted cancer incidence rates (13 oldest SEER registries) declined by 1.8% per year in men and were stable in women, while cancer death rates nationwide decreased by 1.8% per year in men and by 1.4% per year in women. The overall cancer death rate decreased from 215.1 (per 100,000 population) in 1991 to 168.7 in 2011, a total relative decline of 22%. However, the magnitude of the decline varied by state, and was generally lowest in the South (15%) and highest in the Northeast (20%). For example, there were declines of 25% to 30% in Maryland, New Jersey, Massachusetts, New York, and Delaware, which collectively averted 29,000 cancer deaths in 2011 as a result of this progress. Further gains can be accelerated by applying existing cancer control knowledge across all segments of the population. CA Cancer J Clin 2015;000:000000. V C 2015 American Cancer Society.
Article
The purpose of this review is to highlight the recent mechanistic developments elucidating the role of matrix metalloproteinases (MMPs) in tumour invasion and metastasis. The ability of tumour cells to invade, migrate and subsequently metastasise is a fundamental characteristic of cancer. Tumour invasion and metastasis is increasingly being characterised by the dynamic relationship between cancer cells and their microenvironment and developing a greater understanding of these basic pathological mechanisms is crucial. While MMPs have been strongly implicated in these processes as a result of extensive circumstantial evidence; for example increased expression of individual MMPs in tumours and association of specific MMPs with prognosis; the underpinning mechanisms are only now being elucidated. Recent studies are now providing a mechanistic basis, highlighting and reinforcing the catalytic and non-catalytic roles of specific MMPs as key players in tumour invasion and metastasis. This article is protected by copyright. All rights reserved.
Article
Purpose: BMS-275291 is a novel broad-spectrum inhibitor of matrix metalloproteinase (MMPs) rationally designed to spare a class of closely related metalloproteinases known as sheddases. Inadvertent sheddase inhibition is hypothesized to play a role in the dose-limiting joint toxicities occurring with hydroxamate-based MMP inhibitors. This trial was conducted to establish the recommended phase II dose; determine safety, toxicity, and pharmacokinetics of BMS-275291; and to assess potential markers of sheddase activity [tumor necrosis factor-α (TNFα) release and TNFα-RII shedding]. Experimental Design: This was an open label, single arm, phase I study conducted at two centers. Patients with advanced or metastatic cancer were treated with once-daily oral BMS-275291 at doses escalating from 600 to 2400 mg/day. Six to eight patients/dose level were to be studied with the recommended phase II dose level expanded to a total of 15 patients. Pharmacokinetic sampling was performed on days 1, 15, and 29 at 0, 0.5, 1, 2, 4, 6, 8, and 24 h after dosing. Radiological tumor assessment was performed every 8 weeks. Results: Forty-four evaluable patients were enrolled in this study with the most frequent tumor types being colorectal cancer and non-small cell lung cancer. Dose limiting toxicities were observed at 600 mg/day (one of eight patients with grade 3 transaminitis) and at 1200 mg/day (1 of 15 patients with grade 3 rash and grade 4 shortness of breath), both in the context of predisposing conditions. No dose-limiting toxicities occurred at 900, 1800, or 2400 mg/day. The most frequent adverse events considered possibly, probably, or definitely drug-related were joint toxicity (myalgia/arthralgia), rash, fatigue, headache, nausea, and taste change, all of which were mild, grade 1, grade 2, and not dose-limiting. No objective tumor responses were observed. Twelve of forty-four patients received treatment for 4+ months, six for 8+ months, three for >1 year. Desired trough levels of parent BMS-275291 were maintained with once daily dosing. The mean plasma concentration of parent BMS-275291 at trough exceeded the calculated in vitro IC80 value for MMP-2 and IC90 value for MMP-9 at the recommended phase II dose of 1200 mg/day. No major changes in serum concentrations of sheddase enzymatic products, TNFα or TNFα-RII, were observed. Conclusions: BMS-275291 is a nonhydroxamate MMP inhibitor with a novel mercaptoacyl zinc-binding group. In this study, plasma concentrations of BMS-275291 continuously exceeded in vitro MMP IC50 values without dose-limiting joint toxicity. In this refractory patient population, a suggestion of disease stabilization was observed in 12 patients. On the basis of preclinical, clinical, and pharmacokinetic data, the recommended phase II dose for future study is 1200 mg/day.