Abstract
The metzincin clan of metalloproteinases includes the MMP, disintegrin and metalloproteinase (ADAM) and ADAM with thrombospondin motifs families, which cleave extracellular targets in a wide range of (patho)physiological processes. Antibodies constitute a powerful tool to modulate the activity of these enzymes for both therapeutic and research purposes. In this review, we give an overview of monoclonal antibodies (mAbs) that have been tested in preclinical disease models, human trials and important studies of metzincin structure and function. Initial attempts to develop therapeutic small molecule inhibitors against MMPs were hampered by structural similarities between metzincin active sites and, consequently, off‐target effects. Therefore, more recently, mAbs have been developed that do not bind to the active site but bind to surface‐exposed loops that are poorly conserved in closely related family members. Inhibition of protease activity by these mAbs occurs through a variety of mechanisms, including (i) barring access to the active site, (ii) disruption of exosite binding, and (iii) prevention of protease activation. These different modes of inhibition are discussed in the context of the antibodies' potency, selectivity and, importantly, the effects in models of disease and clinical trials. In addition, various innovative strategies that were used to generate anti‐metzincin mAbs are discussed.
Linked Articles
This article is part of a themed section on Translating the Matrix. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.1/issuetoc
Abbreviations
- Cat
catalytic domain
- CysR
cysteine‐rich
- Fab
antigen‐binding fragment
- Fc
crystallizable fragment
- Hpx
haemopexin‐like
- mAb
monoclonal antibody
- MT‐MMP
membrane‐type MMP
- OA
osteoarthritis
- RA
rheumatoid arthritis
- scFv
single‐chain variable fragment
- Sp
spacer
- TIMP
tissue inhibitor of metalloproteinase
- TS
thrombospondin
- TTP
thrombotic thrombocytopenic purpura
- UC
ulcerative colitis
- VH
variable region of the heavy chain
- VL
variable region of the light chain
- VWF
von Willebrand factor
Introduction
Monoclonal antibodies (mAbs) are widely used as drugs and in research. The isotype used is usually immunoglobulin G (IgG). IgGs are tetramers of ~150 kDa, which comprise a pair of identical heavy and light chains linked by disulfide bonds (Figure 1). IgGs are divalent, that is, they contain two antigen binding sites, each composed of the variable regions of the heavy and light chains (VH and VL, respectively). Increasingly, smaller antibody fragments that retain the antigen binding site are used. Single‐chain variable fragment (scFv) comprises the variable regions of the VH and VL of an IgG molecule connected with a short glycine‐rich linker. This fusion protein retains the specificity of IgG and constitutes a minimal antigen binding site. Antigen‐binding fragments (Fabs) are composed of one constant and one variable domain from the heavy and light chains connected by a disulfide bond. scFv can also be expressed as fusion proteins together with the crystallizable fragment (Fc) region of the IgG molecule, which comprises the constant domains (CH) of the heavy chains. The resulting molecule (scFv‐Fc) retains both the antigen binding site, provided by the scFv, and the immune effector functions, provided by the Fc region. The Fc region is recognized by Fc receptors on various immune cells and mediates cytotoxic functions and clearance of immune complexes of mAbs bound to their extracellular target. The combination of high potency and high selectivity for their target has not only made mAbs an important new class of therapeutics but also an invaluable tool in studies of protein function, including that of the metalloproteinase clan called metzincins.
Figure 1.

Schematic illustration of IgG and IgG fragments. IgGs are bivalent and consist of two heavy and two light chains, made of variable (VH and VL) and constant (CH and CL) regions. The heavy chain constant region is divided into CH1, CH2 and CH3. The antigen binding site is composed of six hypervariable complementarity determining regions, three in the variable region of the heavy (VH) and light (VL) chains. The ‘fragment crystallizable’ (Fc) region is composed of the CH2 and CH3 constant regions of the antibody and mediates interactions with cell surface (Fc) receptors and the complement system. The antigen‐binding fragment (Fab) is composed of one constant and one variable region of each of the heavy and light chains (monovalent). It can be produced from an intact IgG by digestion with papain, or it can be expressed recombinantly. The scFv consists of single VH and VL regions connected by a flexible linker and is therefore monovalent. The scFv‐Fc is bivalent and consists of two scFvs connected to the Fc region of IgG. Yellow lines indicate disulfide bonds.
Metzincins are metallopeptidases that share a highly similar catalytic site. This contains three histidines that coordinate a zinc ion and a glutamate, which together with a bound water molecule drive the hydrolysis of the substrate peptide bond. The structure of their catalytic site is further formed by the consensus sequence HEXXHXXG/NXXH/D. This motif is followed C‐terminally by a conserved methionine residue which constitutes a tight turn (named ‘Met‐turn’) and contributes to the structural integrity of the catalytic domain (Cat) (Bode et al., 1993; Tallant et al., 2010). The best known metzincin families are the matrix metalloproteinases (MMPs) and the adamalysins, which are sometimes called reprolysins and include disintegrin and metalloproteinase (ADAM) and ADAM with thrombospondin motifs (ADAMTS) subfamilies. Other metzincin clan members are the astacins, serralysins, snapalysins, leishmanolysins, pappalysins, archaemetzincins and fragilysins (Gomis‐Rüth et al., 2012). In this review, we give an overview of promising mAbs that have been used in human trials, preclinical models and important studies of the structure and function of metzincins.
MMPs
MMPs (matrixins) play key roles in remodelling and turnover of extracellular matrix and are involved in embryonic development, wound healing, angiogenesis, arthritis, cardiovascular diseases and cancer (Murphy and Nagase, 2008). They also cleave intracellular substrates (Ali et al., 2010; Cauwe and Opdenakker, 2010; Jobin et al., 2017) and act as transcriptional activators (Marchant et al., 2014).
The MMP family comprises 23 members, which are generally classified as collagenases (MMP‐1, ‐8 and ‐13), gelatinases (MMP‐2 and MMP‐9), stromelysins (MMP‐3 and ‐10), matrilysins (MMP‐7, ‐11 and ‐26) and membrane‐type MMPs (MT‐MMPs) (MMP‐14, ‐15, ‐16, ‐17, ‐24 and ‐25). They consist of a prodomain, which maintains the zymogen form, a zinc and calcium‐dependent Cat, a linker peptide (hinge region) and a haemopexin‐like (Hpx) domain (Figure 2). Remarkably, the Cat domain of MMP‐2 and ‐9 has fibronectin‐II‐like repeats inserted into a central loop region of the Cat domain, while maintaining the same general fold of the metzincin Cat‐domain.
Figure 2.

Domain organization of metzincins discussed in this review. S, signal peptide; Pro, prodomain; F, fibronectin type II repeats; H, hinge region; HPX, haemopexin domain; TM, transmembrane domain; CT, c‐terminal tail; Dis, disintegrin‐like domain; CR, cysteine‐rich domain; EGF‐L, EGF‐like domain; TS, thrombospondin type‐1‐like repeat; CUB, complement c1r/c1s, uegf, bmp1 domain; LAMG, laminin G‐like domain; ID, interdomain region. For tandem repeats, the number of repeats is shown in brackets. Note that the disintegrin‐like domains of ADAMs and ADAMTSs are structurally very different.
Small molecule inhibitors have been developed that target the catalytic zinc ion of MMPs, but their selectivity is generally poor, which can be explained by the similarity of the active sites of MMPs and metzincins in general (Coussens et al., 2002). MAbs, however, have the advantage that they can be developed to bind variable regions outside the catalytic site, while maintaining inhibitory potential by blocking access to the active site or exosites (Table 1).
Table 1.
Properties of mAbs developed against metzincin targets
| mAb | Format | Target | Epitope | K D (nM) | K i (nM) | Effect | Effective Concentration in vitro/ex vivo (nM) | Effective concentration in vivo (mg·kg−1) | Reference |
|---|---|---|---|---|---|---|---|---|---|
| F2–1‐11 G8–25‐5 | IgG | MMP‐2 | ND | ND | ND | Inhibits catalytic activity | ND | ND | Emara and Wozniak, 2010 |
| SZ‐117 | IgG | MMP‐2 | Fibronectin type 2 | ND | ND | Inhibits catalytic activity | 1000 | ND | Bu et al., 2012 |
| Nb14 | Nb | MMP‐8 | Cat | 0.24‐1.7 | 4400a , b | Inhibits catalytic activity | ND | ND | Demeestere et al., 2016 |
| 19500c | |||||||||
| SDS3 | IgG | MMP‐9 | Catalytic zinc | 200 | 1000 | Inhibits catalytic activity | ND | 1–5 | Sela‐Passwell et al., 2011 |
| SDS4 | IgG | MMP‐9 | Catalytic zinc | 20 | 54 | Inhibits catalytic activity | ND | ND | Sela‐Passwell et al., 2011 |
| 5F3, 6B2, 6D2, 6D7 | IgG | MMP‐9 | ND | ND | ND | Inhibit catalytic activity | 667 | ND | Höyhtya et al., 1990 |
| 4B7 | IgG | MMP‐9 | ND | ND | ND | Increases catalytic activity | 667 | ND | Höyhtya et al., 1990 |
| REGA‐3G12 | IgG | MMP‐9 | Cat | 2.1 | NDb , c | Inhibits catalytic activity | ND | ND | Paemen et al., 1995 |
| Martens et al., 2007 | |||||||||
| scFv | – | – | – | – | – | 16 670 (0.5 mg·mL−1) | 1–2 | Pruijt et al., 1999 Shimanovich et al., 2004 | |
| 6‐6B | IgG | MMP‐9 | ND | ND | Complete inhibition at 1 μg·mL−1 | Inhibits enzyme activation | ND | ND | Ramos‐DeSimone et al., 1993 |
| GS‐5745 | IgG | MMP‐9 | Cat | 0.17 | 0.22a | Inhibits catalytic activity and enzyme activation | ND | 15 | Marshall et al., 2015, g |
| Appleby et al., 2017 | |||||||||
| 0.008–0.04 (pro)3.3 (Cat) | 0.3–1.3a , b | ||||||||
| – | – | – | – | – | – | – | – | 0.3–5 | Sandborn et al., 2016 |
| 14D10 | IgG | MMP‐13 | Active site | 0.33 | 12a 20c | Inhibits catalytic activity | 260 | ND | Naito et al., 2012 |
| DX‐2400 | IgG | MMP‐14 | Cat | ND | 0.8 | Inhibits catalytic activity | 6.7–67 | 10–30 | Devy et al., 2009 |
| ND | 0.2–100 | Ager et al., 2015 | |||||||
| 10–500 | 20–40 | Kaneko et al., 2016 | |||||||
| 3A2 | Fab | MMP‐14 | Cat | 4.8 | 9.7a | Inhibits catalytic activity | 200–250 | ND | Nam et al., 2016 |
| 1F8 | Fab | MMP‐14 | Cat | 8.3 | 110 | Inhibits catalytic activity | ND | ND | Nam et al., 2017 |
| R2C5 | Fab | MMP‐14 | Cat | 15 | 50a | Inhibits catalytic activity | ND | ND | Lopez et al., 2017 |
| R2C7 | Fab | MMP‐14 | Cat | 27 | 100a | Inhibits catalytic activity | ND | ND | Lopez et al., 2017 |
| R2C14 | Fab | MMP‐14 | Cat | 6.1 | 10a | Inhibits catalytic activity | ND | ND | Lopez et al., 2017 |
| E2_C6 A4_7 | scFc‐Fv | MMP‐14 | Cat | 0.11–4 | 100a , b | Inhibits catalytic activity and pro‐MMP‐2 activation | 3–500 | 5 | Botkjaer et al., 2016 |
| 3A2 | Fab | MMP‐14 | Cat | 8 | 18a | Inhibits catalytic activity and pro‐MMP‐2 activation | 200–500 | 10–15 | Remacle et al., 2017 |
| LOOPab | IgG | MMP‐14 | MT‐loop | ND | ND | Inhibits cell localization | 100 | ND | Woskowicz et al., 2013 |
| 9E8 | IgG | MMP‐14 | MT‐loop | ND | 0.6 | Inhibits proMMP‐2 activation | 10–650 | ND | Ingvarsen et al. 2008 Shiryaev et al., 2013 |
| LEM‐2/15 | Fab | MMP‐14 | V‐B loop | 2.3 | 45a | Inhibits catalytic activity | 5000 | ND | Galvez et al., 2001 |
| 3 | Udi et al. Talmi‐Frank et al., 2016 | ||||||||
| Fab | |||||||||
| mAb‐1 | IgG | MMP‐14 | Haemopexin | ND | ND | Induces MMP‐14 dimerization | 330 | ND | Ingvarsen et al., 2008 |
| CHL | scFv | MMP‐14 | Haemopexin | 169 | ND | Inhibits proMMP‐2 activation | 1560–5000 | ND | Basu et al., 2012 |
| CHA | scFv | MMP‐14/15 | Haemopexin | 10.7 | ND | Inhibits proMMP‐2 activation and CD44 shedding | 420–5000 | ND | Basu et al., 2012 |
| Mab1031 | IgG | ADAM8 | Cat/Dis | ND | ND | Inhibits catalytic activity | 133 | 0.5–1.5 | Romagnoli et al., 2014 |
| 8C7 | IgG | ADAM10 | CysR | 93 | ND | Inhibits catalytic activity Inhibits ephrin endocytosis | 667–2670 | ND | Atapattu et al., 2012 |
| D1(A12) | Fab | ADAM17 | Cat/Dis/Cys | 0.46 | 0.89a | Inhibits catalytic activity | 8.6a | – | Tape et al., 2011 |
| – | IgG | – | – | – | 74e | – | 8‐11a | – | Tape et al., 2011 |
| – | IgG | – | – | – | – | – | 200 | 10 | Richards et al., 2012 |
| A9(B8) | IgG | ADAM17 | Cat | 0.33 | 1.19 | Inhibits catalytic activity | 200–250a | ‐10 | Kwok et al., 2014 |
| 200–250 | – | Ye et al., 2017 | |||||||
| A300E‐BiTE | Bispecific scFV | ADAM17/CD‐3 | CysR | ND | ND | Induces T‐cell mediated lysis of cancer cells | 6.67 | ND | Yamamoto et al., 2012 |
| MEDI3622 | IgG | ADAM17 | Cat | 0.039 | 3.1a | Inhibits catalytic activity | 0.9a 400 | 1–30 | Rios‐Doria et al., 2015 Peng et al., 2016 |
| 7E8.1E3 | IgG | ADAMTS‐4 | Cat/Dis | 0.25 | 0.035a , d | Inhibits catalytic activity | 670 | ND | Larkin et al., 2015 |
| 7C7.1H1 | IgG | ADAMTS‐4 | CysRich/Sp | 0.29 | 0.048a , d | Inhibits catalytic activity | 670 | ND | Larkin et al., 2015 |
| GSK2394000 | IgG | ADAMTS‐5 | Cat/Dis | 0.21 | 11a , d | Inhibits catalytic activity | 670 | ND | Larkin et al., 2015 |
| GSK2394002 | IgG | ADAMTS‐5 | Cat/Dis | 0.038 | 0.083a , d | Inhibits catalytic activity | 670 | 10–16 30–300 (Cynomolgus m.) | Larkin et al., 2015 |
| 10 | Miller et al., 2016 | ||||||||
| 2B9 | scFc‐Fv | ADAMTS‐5 | Sp | 6.6 | 90–140a , d | Inhibits activity against macromolecular substrates | ND | ND | Santamaria et al., 2015 |
| 2D3 | scFc‐Fv | ADAMTS‐5 | Cat/Dis | 3.9 | 2.5a | Inhibits catalytic activity | 10–100 | ND | Santamaria et al., 2015 |
| 8–90a , d | |||||||||
| 250 | – | Yamamoto et al., 2017 | |||||||
| 1B7 | scFc‐Fv | ADAMTS‐5 | Cat/Dis | 70 | NI | Inhibits endocytosis | 100 | ND | Santamaria et al., 2017 |
| CRB0017 | IgG | ADAMTS‐5 | Sp | 2.2 | ND | Inhibits aggrecanase activity | 20 | 12 μg/knee | Visintin et al., 2012 |
| Chiusaroli et al., 2013 | |||||||||
| 237–53 | Fab | ADAMTS‐4 | TS‐1 | 12 | 80a , d | Inhibits aggrecanase activity | 5.4 | ND | Shiraishi et al., 2015 |
| 237–53 | Fab | ADAMTS‐5 | TS‐1 | 1.5 | ND | Inhibits aggrecanase activity | 5.4 | ND | Shiraishi et al., 2015 |
| PAC1 PAC2 | scFv | PAPP‐A | LNR3 | 0.25 | 1.2 | Inhibit cleavage of IGFBP‐4 but not IGFBP‐5 | ND | ND | Mikkelsen et al., 2008 |
| 1/41 | IgG | PAPP‐A | LNR3 | 0.097 | 0.14 | Inhibits catalytic activity | 1–0.001 | 10–30 | Mikkelsen et al., 2014 |
| – | – | – | – | – | – | – | – | 30 | Becker et al., 2015 |
| 129 | Fab | ADAMDEC1 | Dis | 9.3 | ND | Inhibit catalytic activity | ND | ND | Lund et al., 2018 |
| 177 | Fab | – | Dis | 1.4 | ND | Increase catalytic activityf | ND | ND | – |
| 111 | Fab | – | Dis | 0.38 | ND | – | ND | ND | – |
Binding constants (K D) were measured by solid‐phase binding assays or surface plasmon resonance. Inhibition constants (K i) were measured, whenever possible, against synthetic peptide substrates. Where the effective concentrations in vitro have been reported in μg·mL−1 in the original papers, they have been converted to molar concentrations.
IC50 value;
measured against gelatin;
measured against collagen;
measured against aggrecan;
measured against TNF‐α;
measured against casein
GS‐5745 is the humanized version of AB0041
ND, not determined, only single concentrations tested; NI, not inhibitory; Nb, nanobody, single variable domain derived from heavy‐chain‐only antibodies of Camelidae.
Gelatinases (MMP‐2 and ‐9)
The gelatinase subfamily consists of MMP‐2 and MMP‐9. MMP‐9 cleaves collagen IV and laminin in the basement membrane, which is thought to affect tumour cell invasion and metastasis (Overall and Kleifeld, 2006). Matrix remodelling by gelatinases also appears to promote the growth of tumour cells, probably by facilitating vascularization (Hu et al., 2007). MMP‐9 has also been implicated in inflammation‐related tissue damage. In this context, it has been studied in inflammatory bowel disease, cystic fibrosis, rheumatoid arthritis (RA) and bullous pemphigoid (Gaggar et al., 2007; Hu et al., 2007; Fanjul‐Fernandez et al., 2010; de Bruyn et al., 2016). MMP‐9 is, therefore, considered a drug target in several of these conditions.
One of the first inhibitory mAbs that was developed against MMPs in the early 1990's (REGA‐3G12) binds the Cat domain of MMP‐9 (Paemen et al., 1995; Martens et al., 2007). It proved very useful a few years later when it was used to elucidate a physiological role of MMP‐9 in haematopoiesis (Pruijt et al., 1999). Later on, it also provided important evidence that suggested a potential benefit of targeting MMP‐9 in bullous pemphigoid (Shimanovich et al., 2004). Interestingly, another early anti‐MMP‐9 mAb (Höyhtya et al., 1990) increased proteolysis of type IV collagen instead of inhibiting it, but unfortunately, the mechanism was not elucidated.
More recently, Sela‐Passwell et al. (2011) immunized mice with a synthetic molecule (a Tris‐imidazole‐zinc complex) that mimics the structure of the zinc‐histidine complex present in the active site of metzincins. They then isolated two mAbs (SDS3 and SDS4) that specifically inhibit MMP‐2 and MMP‐9 but showed no or minimal inhibition of other MMPs and did not bind to free zinc‐ions. However, this strategy does not allow prediction of the metzincin target before immunization and the risk of cross reactivity with other metalloproteinases is high. The crystal structure of the SDS3 Fab‐MMP‐9 Cat domain complex confirmed that the antibody directly bound to the active site zinc through the hydroxyl group of a tyrosine residue in its antigen binding site. Administration of either SDS3 or SDS4 in mouse models of ulcerative colitis (UC) attenuated the severity of the disease, suggesting that targeting of MMP‐2 and MMP‐9 may be a viable therapeutic option in UC.
An anti‐MMP‐9 mAb developed by Gilead Sciences (GS‐5745) recognizes with much higher affinity the proform rather than the active enzyme and inhibits MMP‐9 activation. In addition, it inhibits MMP‐9 catalytic activity directly by ~50%, probably because it sterically hinders access to the active site (Appleby et al., 2017). It was efficacious in preclinical models of UC and cancer (Marshall et al., 2015), and a phase I randomized clinical trial showed that it did not induce musculoskeletal syndrome (Sandborn et al., 2016), a side effect frequently associated with broad spectrum small molecule MMP inhibitors (Coussens et al., 2002). GS‐5745 is currently being tested in several clinical trials under the commercial name of andecaliximab, including a phase 3 trial in patients with gastric cancer (ClinicalTrials gov Identifier NCT02545504) and phase II trials in patients with Crohn's disease (NCT02405442, now completed), RA (NCT02862574) and cystic fibrosis (NCT02759562). A combined phase 2/3 trial in patients with UC (NCT02520284) was stopped due to lack of efficacy.
MMP‐14
MMP‐14 (MT1‐MMP) can digest several extracellular matrix proteins (collagens, fibronectin and laminin), but one of its important roles is the initiation of proteolytic cascades through the activation of other proMMPs such as proMMP‐2 (Sato et al., 1994; Ohuchi et al., 1997). The initial activation of proMMP‐2 requires the formation of a complex between proMMP‐2, tissue inhibitor of metalloproteinase‐2 (TIMP‐2) and an MMP‐14 homo‐dimer. TIMP‐2 inhibits one of the MMP‐14 molecules in the homodimer through its N‐terminal domain whereas its C‐terminal domain binds to the Hpx domain in proMMP‐2. This allows activation of proMMP‐2 by the uninhibited MMP‐14 molecule in the homodimer and subsequent further autocatalytic processing to the fully active form (Itoh, 2015).
Inhibition of MMP‐14 has attracted considerable interest due to its involvement in several pathophysiological processes, including cancer cell invasion, metastasis, angiogenesis (Itoh, 2015), RA (Miller et al., 2009) and lung infection (Talmi‐Frank et al., 2016). The first mAb selectively targeting MMP‐14 was DX‐2400, developed by Dyax Corp (Devy et al., 2009). It was selected from a human Fab phage display library against the Cat domain of MMP‐14 and directly competes with a synthetic peptide substrate. It was shown to be effective in in vitro and in vivo models of angiogenesis, tumour growth and metastasis (Devy et al., 2009; Ager et al., 2015). Moreover, it significantly reduced cartilage degradation and disease progression in a mouse model of RA, where it exerted its effect mainly by reducing the spread of the disease to unaffected joints, with minimal effect on swelling and bone erosion (Kaneko et al., 2016). This study showed the importance of MMP‐14 in cartilage degradation and migration of rheumatoid synovial fibroblasts to unaffected joints. Importantly, the combined inhibition of MMP‐14 and TNF synergistically inhibited both joint damage and synovial inflammation (Kaneko et al., 2016).
Raising conventional antibodies that bind the active site of metzincins is complicated by the fact that the active site is often buried in a cleft that is poorly accessible to the antibodies due to the overall shape of their antigen binding surface (cave‐like, grooved or flat) (Lauwereys et al., 1998). To address this, Nam et al. (2016) introduced the longer camelid complementarity determining region‐H3 into the human antibody scaffold. The idea is that the long, convex‐shaped, camelid‐like paratopes are better able to insert into the active site cleft than conventional antibodies. A human Fab library was then made to isolate inhibitory mAbs against the MMP‐14 Cat. One of the isolated mAbs (Fab 3A2) targets the S1’ pocket of MMP‐14 near the active site zinc and prevents proteolysis of a synthetic peptide substrate.
Targeting the active site, however, has other drawbacks. The active sites of all metzincins are structurally similar, as they all depend on the three histidines, the catalytic glutamic acid residue and a zinc ion. Consequently, mAbs that bind to the active site may suffer from poor selectivity. A solution to this is to target a region outside the active site cleft, which is poorly conserved in close family members. For example, Botkjaer et al. (2016) isolated mAbs that bound to the MMP‐14 Cat domain outside the active site cleft. Therefore, they did not inhibit cleavage of a small peptide substrate but still blocked pro‐MMP‐2 activation, collagen‐film degradation and gelatin‐film degradation, probably due to steric hindrance. The affinity‐matured antibodies were able to inhibit collagen invasion by tumour cells in vitro. They also reduced breast tumour growth and metastasis in a mouse xenograft model (MDA‐MB‐231).
In the MT‐MMPs, a promising strategy is the targeting of the so‐called MT‐loop in the Cat domain (163PYAYIREG170 in MMP‐14) (Figure 3), which lies distant from the active site but contains an exosite that is essential for TIMP‐2 binding and, consequently, MMP‐2 activation (Fernandez‐Catalan et al., 1998; English et al., 2001). It also binds to integrin‐rich cell adhesion complexes, an interaction necessary for locating MMP‐14 to the leading edges of invading cells (Woskowicz et al., 2013). As expected, an antibody against the MT‐loop (EP1264Y) did not prevent proteolysis of a short peptide substrate, gelatin or collagen but reduced cellular invasion (Woskowicz et al., 2013). The MT‐loop is also recognized by another mAb, 9E8, which prevents TIMP‐2 binding and MMP‐2 activation (Ingvarsen et al., 2008). These mAbs show that targeting the MT‐loop can be effective and because the MT‐loop is also present in MMP‐15, ‐16 and ‐24 but differs both in length and sequence, a similar approach can be applied to other MT‐MMPs.
Figure 3.

Structure of the catalytic domain (Cat) of MMP‐14. The Cat of members of the metzincin clan have a characteristic fold consisting of an active site groove that runs approximately along the axis of the central α‐helix beneath it, here shown horizontally at the centre of the domain. This active site helix also provides two of the three histidines that bind the catalytic zinc ion. Structural stability of MMP‐14 is provided by a second zinc ion (purple circle) and two calcium ions (green circles). Indicated are the membrane‐type (MT) loop (residues 163–170) and the VB‐loop (residues 218–233), which have been used for selective targeting by mAbs. PDB ID, 1BQQ (Fernandez‐Catalan et al., 1998).
Besides the MT‐loop, other loops are amenable to selective targeting by mAbs. For example, the V‐B loop (residues 218–233 in MMP‐14; Figure 3) is highly divergent among MMPs. A mAb was generated against the V‐B loop in MMP‐14 by immunization with a cyclic peptide containing the loop sequence (Udi et al., 2015). This mAb (LEM‐2/15) exerts its inhibitory effect by inducing a conformational change in MMP‐14, which results in a narrower substrate‐binding cleft and, consequently, inhibits proteolysis of small synthetic peptides, gelatin and collagen type I. It has, however, only a moderate effect on MMP‐2 activation (Galvez et al., 2001). It also inhibited endothelial cell invasion of collagen and fibrin gels and capillary tube formation in a Matrigel based assay, which highlights the important role of MMP‐14 in angiogenesis (Galvez et al., 2001). Interestingly, LEM‐2/15 was also tested in influenza/Streptococcus pneumonia coinfection models where it showed a synergistic effect with the antiviral drug Tamiflu both in a therapeutic and preventive mode (Talmi‐Frank et al., 2016).
In addition to exosites in the Cat domain, metzincins often depend on exosites in ancillary domains to bind their substrates or co‐factors. Several research labs have therefore targeted ancillary domains to generate function‐specific inhibitory mAbs. The Hpx domain of MMP‐14, for example, is important in unwinding the collagen triple helix for collagenolysis (Tam et al., 2002), as well as proMMP‐2 activation (Itoh et al., 2001; Lehti et al., 2002) and CD44 shedding (Suenaga et al., 2005). The different functions of MMP‐14 rely on distinct exosites, which can be targeted individually. For example, two scFv antibody fragments (named CHL and CHA), isolated against the Hpx domain of MMP‐14, interfere specifically with the collagen‐binding activity without affecting proMMP‐2 activation (Basu et al., 2012). The interference with collagen binding translated into a strong attenuation of cellular invasion in vitro (Basu et al., 2012).
Interestingly, screening of inhibitory mAbs sometimes identifies antibodies that enhance rather than inhibit the function of the target protease. Different mechanisms have been described that explain the ‘activation’. One example is a mAb against the Hpx domain of MMP‐14 that enhances activation of MMP‐2, without affecting collagenolytic activity (Ingvarsen et al., 2008). In this case, the divalent (IgG) antibody supported MMP‐14 dimerization/clustering on the cell surface. A derived Fab (monovalent) was therefore unable to replicate the increase in MMP‐2 activation.
ADAMs
The ADAM family comprises 22 human proteins, 13 of which retain a functional Cat domain (Murphy, 2008). They are membrane‐associated metzincins consisting of a prodomain, a Cat domain, a disintegrin‐like (Dis) domain, a cysteine‐rich (CysR) domain, an EGF‐like domain (except ADAM10 and ADAM17), a transmembrane domain and a cytoplasmic tail (Figure 2). ADAMs are involved in ectodomain shedding of cell surface proteins and play a pivotal role in the extracellular regulation of cellular signalling.
ADAM17
The best characterized ADAM family member is ADAM17, which is also known as TNF‐α converting enzyme. It is involved in inflammation and cancer through its ability to cleave a variety of substrates such as TNF‐α, EGF receptor (EGFR) ligands and Notch1 (Murphy, 2008). The first mAb against ADAM17, D1(A12), was developed using an ingenious two‐step phage display strategy to circumvent cross reactivity with structurally similar MMPs (Tape et al., 2011). The first step was to isolate a VH chain that binds the Cat domain, and then in the second step, a VL chain was selected for binding to the ancillary Dis/CysR region. The idea is that binding to the Cat domain blocks access of substrates to the active site whereas binding to the Dis/CysR region provides specificity. The resulting ‘cross‐domain’ human antibody is a potent inhibitor of ADAM17 and does not inhibit its closest family member, ADAM10 (Tape et al., 2011). Administration of D1(A12) in an animal model of ovarian cancer significantly inhibited tumour growth (Richards et al., 2012). It was also shown to reduce the in vitro proliferation of human breast tumours and squamous cell carcinoma of the head and neck (Huang et al., 2014; Caiazza et al., 2015). Unfortunately, D1(A12) is specific for human ADAM17, which limited its preclinical applications to xenograft models. A second generation mAb, A9(B8), was therefore designed to cross react with mouse ADAM17 (Kwok et al., 2014). This antibody suppressed the development of pancreatic cancer in mice overexpressing oncogenic Kras, by inhibiting both TNF‐α and amphiregulin shedding (Ye et al., 2017). Another anti‐ADAM17 antibody was developed by MedImmune, LLC. This mAb (MED13622) binds to the Cat domain only and acquires its specificity from binding to the highly variable loop between βIV and βV of the five‐stranded β‐sheet (Rios‐Doria et al., 2015; Peng et al., 2016). It effectively inhibited cancer cell growth in the OE21 oesophageal xenograft model, showing an additive effect with cetuximab (anti‐EGFR). MED13622 was also used to ascertain that ADAM17 can drive tumour proliferation through both EGFR‐dependent and independent mechanisms. Moreover, proteomic studies that used this antibody to block shedding by ADAM17 identified new ADAM17 substrates (Rios‐Doria et al., 2015).
Other ADAMs
ADAM8 appears to be predominantly expressed on the surface of various immune cells, where it may act as a sheddase of cell‐adhesion molecules (Gómez‐Gaviro et al., 2007). Although the development of inhibitory mAbs against ADAM8 has not been reported, one commercially available anti‐ADAM8 mAb (Mab1031) was tested for its effect on adhesion/migration of breast cancer cell lines and growth of xenograft breast tumours (Romagnoli et al., 2014). However, the ability of this mAb to inhibit proteolysis by ADAM8 is not known.
ADAM10 was targeted to study its proteolysis of Ephrins, which are membrane‐bound ligands for tyrosine kinases of the ErbB and Eph families. An anti‐ADAM10 mAb (8C7) was generated against a distal substrate binding pocket in the CysR domain of ADAM10 (Atapattu et al., 2012). It was shown to inhibit internalization of (cleaved) ephrin‐A5 and Eph/ephrin‐mediated cell function.
ADAMTS
The ADAMTS family comprises 19 secreted metalloproteinases in humans (Dubail and Apte, 2015). ADAMTSs share a common domain composition consisting of a signal peptide, a prodomain, a metalloproteinase Cat domain, followed by the non‐catalytic ancillary domains; a disintegrin‐like (Dis) domain, a thrombospondin (TS)‐type I motif domain, a CysR domain, a spacer (Sp) domain and a variable number of TS domains in the C‐terminus (with the exception of ADAMTS‐4). Some of the ADAMTSs have further C‐terminal domains (Figure 2).
ADAMTS‐4 and ‐5
ADAMTS‐1, ‐4 and ‐5 cleave proteoglycans such as aggrecan and versican, which play a structural role in many tissues. Aggrecan is a major component of articular cartilage where it provides the ability to resist compressive loads. This function is achieved by creating high osmolarity through hydration of the negatively charged glycosaminoglycans attached to its protein core. Degradation of aggrecan is a clinical hallmark of degenerative joint disorders such as osteoarthritis (OA) and RA. Genetic knockout of ADAMTS‐5 is sufficient to protect mice from cartilage destruction in OA or inflammatory arthritis models (Glasson et al., 2005; Stanton et al., 2005) and ADAMTS‐5 is the most potent aggrecanase in vitro (Gendron et al., 2007). However, siRNA‐mediated knock down of both ADAMTS‐4 and ADAMTS‐5 in human cartilage explants suggested that both enzymes are involved in aggrecan degradation (Song et al., 2007) and both have attracted considerable interest as a therapeutic target in arthritis.
Phage display has been used to obtain mAbs targeting exosites on ADAMTS‐5 (Santamaria et al., 2015). Competition experiments with the endogenous inhibitor of aggrecanases, TIMP‐3, and a small molecule zinc‐chelating inhibitor (GM6001/Ilomastat) showed that all the isolated inhibitory mAbs recognized epitopes outside the active site cleft. One of these (2D3) targets the Cat/Dis domain and strongly inhibited aggrecanase activity both in unstimulated human chondrocyte cultures from healthy donors (Santamaria et al., 2015) and in OA cartilage explants (Yamamoto et al., 2017).
Interestingly, a mAb (1B7) that binds to the ADAMTS‐5 Cat/Dis region did not inhibit its aggrecanolytic activity but blocked the endocytosis of ADAMTS‐5 by LDL receptor‐related protein‐1. This caused human chondrocytes to accumulate ADAMTS‐5, which increased the degradation of exogenously added aggrecan (Santamaria et al., 2017).
The Sp domain of ADAMTS‐5 is known to be important for aggrecan cleavage (Gendron et al., 2007), and this was confirmed by the inhibitory effect on aggrecan cleavage shown by a mAb (2B9) that specifically binds to the Sp domain (Santamaria et al., 2015). An anti‐ADAMTS‐5 mAb developed by Rottapharm (CRB0017), which was raised against an Sp‐domain sequence, was also effective in a mouse model of OA when administered therapeutically (intra‐articular route) (Chiusaroli et al., 2013).
Another mAb, 237‐53, recognizes a common epitope in the TS‐1 domain of both ADAMTS‐4 and ‐5 and inhibited proteolysis of aggrecan in conditioned media of chondrocyte explants (Shiraishi et al., 2015). GlaxoSmithKline (GSK) has also raised several mAbs in mice against these two aggrecanases and tested their efficacy in preclinical models (Larkin et al., 2015). The two humanized anti‐ADAMTS‐5 mAbs, GSK2394002 and GSK2394000, recognized an epitope spanning the Cat and Dis domains, whereas of the two anti‐ADAMTS‐4 mAbs one bound to both the Cat and Dis domain and the other one to an epitope spanning the CysR and Sp domains. These mAbs have been extensively studied in the context of cartilage degradation using human OA explants (Larkin et al., 2015). The anti‐ADAMTS‐5 mAbs effectively inhibited aggrecan cleavage in unstimulated cultures, whereas the anti‐ADAMTS‐4 mAbs had a modest effect. However, both anti‐ADAMTS‐4 and ‐5 mAbs were effective in cytokine‐stimulated cultures (Larkin et al., 2015). These data complement previous findings showing that ADAMTS‐4 expression is induced under inflammatory conditions, whereas that of ADAMTS‐5 is constitutive (Bau et al., 2002; Song et al., 2007; Shiraishi et al., 2015). Prophylactic, systemic treatment with anti‐ADAMTS‐5 mAbs imparted protection from cartilage degradation in a mouse OA model, thus phenocopying genetic deletion of Adamts‐5 (Miller et al., 2016). One mAb (GSK2394002) also decreased aggrecan degradation in cynomolgus monkeys that were pre‐screened for elevated circulating levels of aggrecan fragments (Larkin et al., 2015). However, this later study also highlighted potential challenges posed by ADAMTS‐5 inhibition. An observation of subendocardial haemorrhage prompted monitoring of cardiovascular function, which showed a sustained increase in mean arterial pressure upon >3 mg·kg−1 dosing and ST segment elevation in electrocardiograms upon dosing of >30 mg·kg−1 (Larkin et al., 2014). These cardiovascular side effects of ADAMTS‐5 inhibition may be mediated by modulation of the important physiological role of ADAMTS‐5 as a versicanase in the cardiovascular system (Dupuis et al., 2011).
ADAMTS‐13: lessons from autoantibodies.
ADAMTS‐13 is a component of blood where it proteolyses ultra long (UL) Von Willebrand factor (VWF) multimers that are released into the circulation by endothelial cells. In patients with the autoimmune form of thrombotic thrombocytopenic purpura (TTP), autoantibodies against ADAMTS‐13 lower ADAMTS‐13 levels and activity, which causes UL VWF multimers to remain in the circulation. UL VWF tend to spontaneously aggregate platelets, leading to thrombi that occlude small vessels.
Although ADAMTS‐13 inhibition in vivo is detrimental, we include ADAMTS‐13 in this review because extensive characterizations of antibodies against ADAMTS‐13 have been instrumental in the understanding of ADAMTS‐13 structure/function and its role in disease. Importantly, these findings may be instructive in the development of inhibitory antibodies against other metzincins, other ADAMTS family members in particular.
Extensive epitope mapping of patient autoantibodies has revealed that the CysR/Sp region of ADAMTS‐13 is almost always targeted, whereas other domains are antigenic targets in some patients but not in others (Klaus et al., 2004; Luken et al., 2005; Zheng et al., 2010). Importantly, many patients had no antibodies against the Cat domain, highlighting the potency of antibodies against the ancillary domains. Despite these epitopes being very remote from the active site, the antibodies inhibit ADAMTS‐13 activity directly by preventing ADAMTS‐13 exosites in its ancillary domains to bind VWF (de Groot et al., 2009; 2015; Pos et al., 2010; Thomas et al., 2015). However, it is important to note that this interference with substrate binding is accompanied by autoantibody‐mediated clearance of ADAMTS‐13, which greatly reduces the ADAMTS‐13 concentration in plasma of TTP patients (Thomas et al., 2015). The precise mechanism by which ADAMTS‐13 antigen/antibody immune complexes are cleared is unknown but may involve Fc receptors on circulating macrophages or Kupffer cells.
A common epitope for autoantibodies in the Sp domain was shown to consist of two arginines, two tyrosines and a phenylalanine provided by neighbouring loop regions (Pos et al., 2011). Interestingly, these residues also appear to form an important exosite that binds VWF (Pos et al., 2010). Nature, therefore, seems to have provided us with a good illustration of inhibitory antibodies that target a distant exosite in an ancillary domain. The amino acids that make up the exosite/epitope are, as expected, poorly conserved in other ADAMTS family members, conferring specificity to both ADAMTS‐13 and the autoantibodies. Consequently, for those ADAMTS family members that are drug targets, these loops in the Sp domain may be a good part of the molecule to target.
Paradoxically, one TTP patient has been described whose high‐titre autoantibodies activated recombinant ADAMTS‐13 threefold when assayed with a VWF fragment (Muia et al., 2014). Although the development of TTP in this patient can be explained by clearance of ADAMTS‐13, the activating effect of the autoantibodies remained intriguing. Various mAbs have since been reported that enhance ADAMTS‐13 activity. Out of the 31 anti‐ADAMTS‐13 mAbs that were screened, 11 showed a modest increase (≤2‐fold) in VWF proteolysis, with some of them showing an additive effect when used in combination (≤4‐fold) (Muia et al., 2014; Deforche et al., 2015). The activating mAbs bind to epitopes on the C‐terminal domains of ADAMTS‐13 (TSP5‐CUB, Figure 2). In vitro, these C‐terminal domains appear to shield an exosite, which is exposed when mAbs (or VWF C‐terminal domains) bind and alter the ADAMTS‐13 conformation (Muia et al., 2014).
Other metzincins
Pappalysin‐1, also known as pregnancy‐associated plasma protein‐A (PAPP‐A) cleaves both insulin‐like growth factor‐binding protein 4 (IGFBP‐4) and ‐5, thus regulating insulin growth factor bioavailability. It consists of a laminin G‐like module, Cat domain, five complement control protein (CCP) modules and three Lin12‐Notch repeats (LNR) of which two are inserted into the Cat domain and a third C‐terminal to the CCP modules (Figure 2). Interestingly, an scFv against the C‐terminal LNR domain of PAPP‐A inhibited proteolysis of IGFBP‐4 much better than that of IGFBP‐5 (Mikkelsen et al., 2008). This highlights the potential of mAbs against exosites in the ancillary domains to differentially inhibit cleavage of a particular substrate. A mAb with a similar epitope (mAb‐PA) also inhibited tumour growth in lung and ovarian tumour xenografts (Mikkelsen et al., 2014; Becker et al., 2015).
Recently, mAbs against decysin‐1 (ADAMDEC1), an ADAM‐like metzincin with unknown function, have been reported (Lund et al., 2018). ADAMDEC‐1 consists only of a Cat domain followed by a short Dis domain (Mueller et al., 1997). Unusually, its catalytic zinc ion is coordinated by two histidines and an aspartic acid residue, rather than three histidine residues (Mueller et al., 1997). It was shown that mAbs targeting the Dis domain can have either an inhibitory or stimulating effect on caseinolytic activity (Lund et al., 2018), suggesting that even very short metzincins such as ADAMDEC1 are amenable to mAb‐mediated ‘activation’.
Conclusions and future directions
MAbs have an instrumental role in dissecting the functions of metzincins in biological systems and have great potential as therapeutic inhibitors in several diseases. As therapeutic inhibitors, mAbs have advantages and disadvantages compared to small molecule inhibitors. The major advantage of mAbs is their very high degree of selectivity, which has proven very difficult to achieve with small molecule inhibitors of metzincins. Other advantages are that metabolic drug–drug interactions are rare and toxicity is generally low. Compared to small molecule inhibitors, most mAbs will also cover a larger surface contact area, which makes them more suited to disrupt high affinity interactions between metzincins and their substrates/cofactors through direct competition or steric hindrance.
A major drawback of therapeutic mAbs in general is that they need to be injected, either subcutaneously or intravenously. Moreover, their usage is restricted to extracellular targets. Although metzincins operate mostly extracellularly, it was recently discovered that several MMPs are active intracellularly (Ali et al., 2010; Jobin et al., 2017). The inability of mAbs to reach the cytoplasm could potentially explain a lack of efficacy of mAbs in some in vivo studies. Specific targeting of intracellular activities of MMPs could potentially be achieved using transfection with antibody constructs that are modified for intracellular localization (‘intrabodies’) (Stocks, 2005; Marschall et al., 2015).
An important consideration for future mAb development is that various metzincins use exosites (in Cat and ancillary domains) to bind co‐factors and substrates, which can therefore be targeted to inhibit the enzyme. Several examples of this have been given in this review (e.g. MMP‐14, ADAM17 and ADAMTS‐5). Autoantibodies against ADAMTS‐13 also highlight the inhibitory potential of antibodies against ancillary domains through both disruption of substrate binding and clearance of immune complexes. For those future metzincin targets that have multiple physiological substrates, substrate‐specific inhibition may be achieved by mAbs against exosites that mediate susbtrate specificity. Examples of such mAbs have already been described (e.g. Mikkelsen et al., 2008). Here lies also an important task for metzincin research, that is, to identify the exosite(s) that dictate substrate specificity.
So far, the development of mAbs has been limited to few metzincin targets (Table 1), and Gilead Science's anti‐MMP‐9 mAb, GS‐5745, appears currently the only anti‐metzincin mAb tested in clinical trials. As research progresses and more becomes known about the roles that metzincins play in various diseases, this will change. There are already several metzincins with known roles in disease, which may be future targets (Overall and Kleifeld, 2006; Yang et al., 2017). A prominent example is ADAMTS‐7, which is emerging as a potential target in cardiovascular disease (Arroyo and Andrés, 2015).
In summary, there is a lot of potential for the development of highly specific mAb inhibitors against MMPs, ADAMTSs and ADAMSs. The increasing use of therapeutic mAbs in general suggests that the expansion of this field will ultimately lead to anti‐metzincin mAbs able to modify detrimental roles in disease.
Nomenclature of targets and ligands
Key protein targets and ligands in this article are hyperlinked to corresponding entries in http://www.guidetopharmacology.org, the common portal for data from the IUPHAR/BPS Guide to PHARMACOLOGY (Harding et al., 2018), and are permanently archived in the Concise Guide to PHARMACOLOGY 2017/18 (Alexander et al., 2017).
Conflict of interest
The authors declare no conflicts of interest.
Acknowledgements
S.S. is supported by the Wellcome Trust Institutional Strategic Support Fund's Faculty Fellowship Scheme (ISSF), Imperial College London.
Santamaria, S. , and de Groot, R. (2019) Monoclonal antibodies against metzincin targets. British Journal of Pharmacology, 176: 52–66. 10.1111/bph.14186.
References
- Ager EI, Kozin SV, Kirkpatrick ND, Seano G, Kodack DP, Askoxylakis V et al (2015). Blockade of MMP14 activity in murine breast carcinomas: implications for macrophages, vessels, and radiotherapy. J Natl Cancer Inst 107 10.1093/jnci/djv017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander SPH, Fabbro D, Kelly E, Marrion NV, Peters JA, Faccenda E et al (2017). The Concise Guide to PHARMACOLOGY 2017/18: Enzymes. 174: S272–S359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ali MA, Cho WJ, Hudson B, Kassiri Z, Granzier H, Schulz R (2010). Titin is a target of matrix metalloproteinase‐2: implications in myocardial ischemia/reperfusion injury. Circulation 122: 2039–2047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Appleby TC, Greenstein AE, Hung M, Liclican A, Velasquez M, Villaseñor AG et al (2017). Biochemical characterization and structure determination of a potent, selective antibody inhibitor of human MMP9. J Biol Chem 292: 6810–6820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arroyo AG, Andrés V (2015). ADAMTS7 in cardiovascular disease: from bedside to bench and back again? Circulation 131: 1156–1159. [DOI] [PubMed] [Google Scholar]
- Atapattu L, Saha N, Llerena C, Vail ME, Scott AM, Nikolov DB et al (2012). Antibodies binding the ADAM10 substrate recognition domain inhibit Eph function. J Cell Sci 125 (Pt 24): 6084–6093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Basu B, Correa de Sampaio P, Mohammed H, Fogarasi M, Corrie P, Watkins NA et al (2012). Inhibition of MT1‐MMP activity using functional antibody fragments selected against its hemopexin domain. Int J Biochem Cell Biol 44: 393–403. [DOI] [PubMed] [Google Scholar]
- Bau B, Gebhard PM, Haag J, Knorr T, Bartnik E, Aigner T (2002). Relative messenger RNA expression profiling of collagenases and aggrecanases in human articular chondrocytes in vivo and in vitro . Arthritis Rheum 46: 2648–2657. [DOI] [PubMed] [Google Scholar]
- Becker MA, Haluska P Jr, Bale LK, Oxvig C, Conover CA (2015). A novel neutralizing antibody targeting pregnancy‐associated plasma protein‐a inhibits ovarian cancer growth and ascites accumulation in patient mouse tumorgrafts. Mol Cancer Ther 14: 973–981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bode W, Gomis‐Rüth FX, Stöcker W (1993). Astacins, serralysins, snake venom and matrix metalloproteinases exhibit identical zinc‐binding environments (HEXXHXXGXXH and Met‐turn) and topologies and should be grouped into a common family, the ‘metzincins’. FEBS Lett 331: 134–140. [DOI] [PubMed] [Google Scholar]
- Botkjaer KA, Kwok HF, Terp MG, Karatt‐Vellatt A, Santamaria S, McCafferty J et al (2016). Development of a specific affinity‐matured exosite inhibitor to MT1‐MMP that efficiently inhibits tumor cell invasion in vitro and metastasis in vivo . Oncotarget 7: 16773–16792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bu Z, Pan Y, Shang B, Cao Z, Zhou Q, Ruan C (2012). SZ‐117, a monoclonal antibody against matrix metalloproteinase‐2 inhibits tumor cell‐mediated angiogenesis. Hybridoma 31: 63–67. [DOI] [PubMed] [Google Scholar]
- Caiazza F, McGowan PM, Mullooly M, Murray A, Synnott N, O'Donovan N et al (2015). Targeting ADAM‐17 with an inhibitory monoclonal antibody has antitumour effects in triple‐negative breast cancer cells. Br J Cancer 112: 1895–1903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cauwe B, Opdenakker G (2010). Intracellular substrate cleavage: a novel dimension in the biochemistry, biology and pathology of matrix metalloproteinases. Crit Rev Biochem Mol Biol 45: 351–423. [DOI] [PubMed] [Google Scholar]
- Chiusaroli R, Visentini M, Galimberti C, Casseler C, Mennuni L, Covaceuszach S et al (2013). Targeting of ADAMTS‐5's ancillary domain with the recombinant mAb CRB0017 ameliorates disease progression in a spontaneous murine model of osteoarthritis. Osteoarthritis Cartilage 21: 1807–1810. [DOI] [PubMed] [Google Scholar]
- Coussens LM, Fingleton B, Matrisian LM (2002). Matrix metalloproteinase inhibitors and cancer: trials and tribulations. Science 295: 2387–2392. [DOI] [PubMed] [Google Scholar]
- Deforche L, Roose E, Vandenbulcke A, Vandeputte N, Feys HB, Springer TA et al (2015). Linker regions and flexibility around the metalloprotease domain account for conformational activation of ADAMTS‐13. J Thromb Haemost 13: 2063–2075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Bruyn M, Vandooren J, Ugarte‐Berzal E, Arijs I, Vermeire S, Opdenakker G (2016). The molecular biology of matrix metalloproteinases and tissue inhibitors of metalloproteinases in inflammatory bowel diseases. Crit Rev Biochem Mol Biol 51: 295–358. [DOI] [PubMed] [Google Scholar]
- de Groot R, Bardhan A, Ramroop N, Lane DA, Crawley JT (2009). Essential role of the disintegrin‐like domain in ADAMTS13 function. Blood 113: 5609–5616. [DOI] [PubMed] [Google Scholar]
- de Groot R, Lane DA, Crawley JT (2015). The role of the ADAMTS13 cysteine‐rich domain in VWF binding and proteolysis. Blood 125: 1968–1975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Demeestere D, Dejonckheere E, Steeland S, Hulpiau P, Haustraete J, Devoogdt N et al (2016). Development and validation of a small single‐domain antibody that effectively inhibits matrix metalloproteinase 8. Mol Ther 24: 890–902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Devy L, Huang L, Naa L, Yanamandra N, Pieters H, Frans N et al (2009). Selective inhibition of matrix metalloproteinase‐14 blocks tumor growth, invasion, and angiogenesis. Cancer Res 69: 1517–1526. [DOI] [PubMed] [Google Scholar]
- Dubail J, Apte SS (2015). Insights on ADAMTS proteases and ADAMTS‐like proteins from mammalian genetics. Matrix Biol 44‐46: 24–37. [DOI] [PubMed] [Google Scholar]
- Dupuis LE, McCulloch DR, McGarity JD, Bahan A, Wessels A, Weber D et al (2011). Altered versican cleavage in ADAMTS5 deficient mice; A novel etiology of myxomatous valve disease. Dev Biol 357: 152–164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Emara M, Wozniak M (2010). In vitro inhibition of human MMP‐2 collagenolytic and gelatinolytic activity by neutralizing monoclonal antibodies. Immunol Invest 39: 833–848. [DOI] [PubMed] [Google Scholar]
- English WR, Holtz B, Vogt G, Knauper V, Murphy G (2001). Characterization of the role of the ‘MT‐loop’: an eight‐amino acid insertion specific to progelatinase A (MMP2) activating membrane‐type matrix metalloproteinases. J Biol Chem 276: 42018–42026. [DOI] [PubMed] [Google Scholar]
- Fanjul‐Fernandez M, Folgueras AR, Cabrera S, Lopez‐Otin C (2010). Matrix metalloproteinases: evolution, gene regulation and functional analysis in mouse models. Biochim Biophys Acta 1803: 3–19. [DOI] [PubMed] [Google Scholar]
- Fernandez‐Catalan C, Bode W, Huber R, Turk D, Calvete JJ, Lichte A et al (1998). Crystal structure of the complex formed by the membrane type 1‐matrix metalloproteinase with the tissue inhibitor of metalloproteinases‐2, the soluble progelatinase A receptor. EMBO J 17: 5238–5248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gaggar A, Li Y, Weathington N, Winkler M, Kong M, Jackson P et al (2007). Matrix metalloprotease‐9 dysregulation in lower airway secretions of cystic fibrosis patients. Am J Physiol Lung Cell Mol Physiol 293: L96–L104. [DOI] [PubMed] [Google Scholar]
- Galvez BG, Matıas‐Roman S, Albar JP, Sanchez‐Madrid F, Arroyo AG (2001). Membrane type 1‐matrix metalloproteinase is activated during migration of human endothelial cells and modulates endothelial motility and matrix remodeling. J Biol Chem 276: 37491–37500. [DOI] [PubMed] [Google Scholar]
- Gendron C, Kashiwagi M, Lim NH, Enghild JJ, Thogersen IB, Hughes C et al (2007). Proteolytic activities of human ADAMTS‐5. Comparative studies with ADAMTS‐4. J Biol Chem 282: 18294–18306. [DOI] [PubMed] [Google Scholar]
- Glasson SS, Askew R, Sheppard B, Carito B, Blanchet T, Ma HL et al (2005). Deletion of active ADAMTS5 prevents cartilage degradation in a murine model of osteoarthritis. Nature 434: 644–648. [DOI] [PubMed] [Google Scholar]
- Gómez‐Gaviro M, Domínguez‐Luis M, Canchado J, Calafat J, Janssen H, Lara‐Pezzi E et al (2007). Expression and regulation of the metalloproteinase ADAM‐8 during human neutrophil pathophysiological activation and its catalytic activity on L‐selectin shedding. J Immunol 178: 8053–8063. [DOI] [PubMed] [Google Scholar]
- Gomis‐Rüth FX, Botelho TO, Bode W (2012). A standard orientation for metallopeptidases. Biochim Biophys Acta 1824: 157–163. [DOI] [PubMed] [Google Scholar]
- Harding SD, Sharman JL, Faccenda E, Southan C, Pawson AJ, Ireland S et al (2018). The IUPHAR/BPS Guide to PHARMACOLOGY in 2018: updates and expansion to encompass the new guide to IMMUNOPHARMACOLOGY. Nucl Acids Res 46: D1091–D1106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Höyhtya M, Hujanen E, Turpeenniemi‐Hujanen T, Thorgeirsson U, Liotta LA, Tryggvason K (1990). Modulation of type‐IV collagenase activity and invasive behavior of metastatic human melanoma (A2058) cells in vitro by monoclonal antibodies to type‐IV collagenase. Int J Cancer 46: 282–286. [DOI] [PubMed] [Google Scholar]
- Hu J, Van den Steen PE, Sang QX, Opdenakker G (2007). Matrix metalloproteinase inhibitors as therapy for inflammatory and vascular diseases. Nat Rev Drug Discov 6: 480–498. [DOI] [PubMed] [Google Scholar]
- Huang Y, Benaich N, Tape C, Kwok HF, Murphy G (2014). Targeting the sheddase activity of ADAM17 by an anti‐ADAM17 antibody D1(A12) inhibits head and neck squamous cell carcinoma cell proliferation and motility via blockage of bradykinin induced HERs transactivation. Int J Biol Sci 10: 702–714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ingvarsen S, Madsen DH, Hillig T, Lund LR, Holmbeck K, Behrendt N et al (2008). Dimerization of endogenous MT1‐MMP is a regulatory step in the activation of the 72‐kDa gelatinase MMP‐2 on fibroblasts and fibrosarcoma cells. Biol Chem 389: 943–953. [DOI] [PubMed] [Google Scholar]
- Itoh Y, Takamura A, Ito N, Maru Y, Sato H, Suenaga N et al (2001). Homophilic complex formation of MT1‐MMP facilitates proMMP‐2 activation on the cell surface and promotes tumor cell invasion. EMBO J 20: 4782–4793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Itoh Y (2015). Membrane‐type matrix metalloproteinases: their functions and regulations. Matrix Biol 44‐46: 207–223. [DOI] [PubMed] [Google Scholar]
- Jobin PG, Butler GS, Overall CM (2017). New intracellular activities of matrix metalloproteinases shine in the moonlight. Biochim Biophys Acta 1864 (11 Pt A): 2043–2055. [DOI] [PubMed] [Google Scholar]
- Kaneko K, Williams RO, Dransfield DT, Nixon AE, Sandison A, Itoh Y (2016). Selective inhibition of membrane type 1 matrix metalloproteinase abrogates progression of experimental inflammatory arthritis: synergy with tumor necrosis factor blockade. Arthritis Rheumatol 68: 521–531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Klaus C, Plaimauer B, Studt JD, Dorner F, Lämmle B, Mannucci PM et al (2004). Epitope mapping of ADAMTS13 autoantibodies in acquired thrombotic thrombocytopenic purpura. Blood 103: 4514–4519. [DOI] [PubMed] [Google Scholar]
- Kwok HF, Botkjaer KA, Tape CJ, Huang Y, McCafferty J, Murphy G (2014). Development of a ‘mouse and human cross‐reactive’ affinity‐matured exosite inhibitory human antibody specific to TACE (ADAM17) for cancer immunotherapy. Protein Eng Des Sel 27: 179–190. [DOI] [PubMed] [Google Scholar]
- Larkin J, Lohr T, Elefante L, Shearin J, Matico R, Su JL et al (2014). The highs and lows of translational drug development: antibody‐mediated inhibition of ADAMTS‐5 for osteoarthritis disease modification. Osteoarthritis Cartilage 22: S883. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Larkin J, Lohr TA, Elefante L, Shearin J, Matico R, Su JL et al (2015). Translational development of an ADAMTS‐5 antibody for osteoarthritis disease modification. Osteoarthritis Cartilage 23: 1254–1266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lauwereys M, Arbabi Ghahroudi M, Desmyter A, Kinne J, Hölzer W, De Genst E et al (1998). Potent enzyme inhibitors derived from dromedary heavy‐chain antibodies. EMBO J 17: 3512–3520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lehti K, Lohi J, Juntunen MM, Pei D, Keski‐Oja J (2002). Oligomerization through hemopexin and cytoplasmic domains regulates the activity and turnover of membrane‐type 1 matrix metalloproteinase. J Biol Chem 277: 8440–8448. [DOI] [PubMed] [Google Scholar]
- Lopez T, Nam DH, Kaihara E, Mustafa Z, Ge X (2017). Identification of highly selective MMP‐14 inhibitory Fabs by deep sequencing. Biotechnol Bioeng 114: 1140–1150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luken BM, Turenhout EA, Hulstein JJ, Van Mourik JA, Fijnheer R, Voorberg J (2005). The spacer domain of ADAMTS13 contains a major binding site for antibodies in patients with thrombotic thrombocytopenic purpura. Thromb Haemost 93: 267–274. [DOI] [PubMed] [Google Scholar]
- Lund J, Elimar Bitsch AM, Grønbech Rasch M, Enoksson M, Troeberg L, Nagase H et al (2018). Monoclonal antibodies targeting the disintegrin‐like domain of ADAMDEC1 modulates the proteolytic activity and enables quantification of ADAMDEC1 protein in human plasma. MAbs 10: 118–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marchant DJ, Bellac CL, Moraes TJ, Wadsworth SJ, Dufour A, Butler GS et al (2014). A new transcriptional role for matrix metalloproteinase‐12 in antiviral immunity. Nat Med 20: 493–502. [DOI] [PubMed] [Google Scholar]
- Marschall AL, Dübel S, Böldicke T (2015). Specific in vivo knockdown of protein function by intrabodies. MAbs 7: 1010–1035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marshall DC, Lyman SK, McCauley S, Kovalenko M, Spangler R, Liu C et al (2015). Selective allosteric inhibition of MMP9 is efficacious in preclinical models of ulcerative colitis and colorectal cancer. PLoS One 10: e0127063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martens E, Leyssen A, Van Aelst I, Fiten P, Piccard H, Hu J et al (2007). A monoclonal antibody inhibits gelatinase B/MMP‐9 by selective binding to part of the catalytic domain and not to the fibronectin or zinc binding domains. Biochim Biophys Acta 1770: 178–186. [DOI] [PubMed] [Google Scholar]
- Mikkelsen JH, Gyrup C, Kristensen P, Overgaard MT, Poulsen CB, Laursen LS et al (2008). Inhibition of the proteolytic activity of pregnancy‐associated plasma protein‐A by targeting substrate exosite binding. J Biol Chem 283: 16772–16780. [DOI] [PubMed] [Google Scholar]
- Mikkelsen JH, Resch ZT, Kalra B, Savjani G, Kumar A, Conover CA et al (2014). Indirect targeting of IGF receptor signaling in vivo by substrate‐selective inhibition of PAPP‐A proteolytic activity. Oncotarget 5: 1014–1025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miller MC, Manning HB, Jain A, Troeberg L, Dudhia J, Essex D et al (2009). Membrane type 1 matrix metalloproteinase is a crucial promoter of synovial invasion in human rheumatoid arthritis. Arthritis Rheum 60: 686–697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miller RE, Tran PB, Ishihara S, Larkin J, Malfait AM (2016). Therapeutic effects of an anti‐ADAMTS‐5 antibody on joint damage and mechanical allodynia in a murine model of osteoarthritis. Osteoarthritis Cartilage 24: 299–306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mueller CGF, Rissoan M, Salinas B, Ait‐Yahia S, Ravel O, Bridon J et al (1997). Polymerase chain reaction selects a novel disintegrin proteinase from CD40‐activated germinal center dendritic cells. J Exp Med 187: 655–663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Muia J, Zhu J, Gupta G, Haberichter SL, Friedman KD, Feys HB et al (2014). Allosteric activation of ADAMTS13 by von Willebrand factor. Proc Natl Acad Sci U S A 111: 18584–18589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Murphy G (2008). The ADAMs: signalling scissors in the tumour microenvironment. Nat Rev Cancer 8: 929–941. [DOI] [PubMed] [Google Scholar]
- Murphy G, Nagase H (2008). Progress in matrix metalloproteinase research. Mol Aspects Med 29: 290–308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Naito S, Takahashi T, Onoda J, Yamauchi A, Kawai T, Kishino J et al (2012). Development of a neutralizing antibody specific for the active form of matrix metalloproteinase‐13. Biochemistry 51: 8877–8884. [DOI] [PubMed] [Google Scholar]
- Nam DH, Rodriguez C, Remacle AG, Strongin AY, Ge X (2016). Active‐site MMP‐selective antibody inhibitors discovered from convex paratope synthetic libraries. Proc Natl Acad Sci U S A 113: 14970–14975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nam DH, Fang K, Rodriguez C, Lopez T, Ge X (2017). Generation of inhibitory monoclonal antibodies targeting matrix metalloproteinase‐14 by motif grafting and CDR optimization. Protein Eng Des Sel 30: 113–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ohuchi E, Imai K, Fujii Y, Sato H, Seiki M, Okada Y (1997). Membrane type 1 matrix metalloproteinase digests interstitial collagens and other extracellular matrix macromolecules. J Biol Chem 272: 2446–2451. [DOI] [PubMed] [Google Scholar]
- Overall CM, Kleifeld O (2006). Tumour microenvironment – opinion: validating matrix metalloproteinases as drug targets and anti‐targets for cancer therapy. Nat Rev Cancer 6: 227–239. [DOI] [PubMed] [Google Scholar]
- Paemen L, Martens E, Masure S, Opdenakker G (1995). Monoclonal antibodies specific for natural human neutrophil gelatinase B used for affinity purification, quantitation by two‐site ELISA and inhibition of enzymatic activity. Eur J Biochem 234: 759–765. [DOI] [PubMed] [Google Scholar]
- Peng L, Cook K, Xu L, Cheng L, Damschroder M, Gao C et al (2016). Molecular basis for the mechanism of action of an anti‐TACE antibody. MAbs 8: 1598–1605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pos W, Crawley JT, Fijnheer R, Voorberg J, Lane DA, Luken BM (2010). An autoantibody epitope comprising residues R660, Y661, and Y665 in the ADAMTS13 spacer domain identifies a binding site for the A2 domain of VWF. Blood 115: 1640–1649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pos W, Sorvillo N, Fijnheer R, Feys HB, Kaijen PH, Vidarsson G et al (2011). Residues Arg568 and Phe592 contribute to an antigenic surface for anti‐ADAMTS13 antibodies in the spacer domain. Haematologica 96: 1670–1677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pruijt JF, Fibbe WE, Laterveer L, Pieters RA, Lindley IJ, Paemen L et al (1999). Prevention of interleukin‐8‐induced mobilization of hematopoietic progenitor cells in rhesus monkeys by inhibitory antibodies against the metalloproteinase gelatinase B (MMP‐9). Proc Natl Acad Sci U S A 96: 10863–10868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ramos‐DeSimone N, Moll UM, Quigley JP, French DL (1993). Inhibition of matrix metalloproteinase 9 activation by a specific monoclonal antibody. Hybridoma 12: 349–363. [DOI] [PubMed] [Google Scholar]
- Remacle AG, Cieplak P, Nam DH, Shiryaev SA, Ge X, Strongin AY (2017). Selective function‐blocking monoclonal human antibody highlights the important role of membrane type‐1 matrix metalloproteinase (MT1‐MMP) in metastasis. Oncotarget 8: 2781–2799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Richards FM, Tape CJ, Jodrell DI, Murphy G (2012). Anti‐tumour effects of a specific anti‐ADAM17 antibody in an ovarian cancer model in vivo . PLoS One 7: e40597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rios‐Doria J, Sabol D, Chesebrough J, Stewart D, Xu L, Tammali R et al (2015). A monoclonal antibody to ADAM17 inhibits tumor growth by inhibiting EGFR and non‐EGFR‐mediated pathways. Mol Cancer Ther 14: 1637–1649. [DOI] [PubMed] [Google Scholar]
- Romagnoli M, Mineva ND, Polmear M, Conrad C, Srinivasan S, Loussouarn D et al (2014). ADAM8 expression in invasive breast cancer promotes tumor dissemination and metastasis. EMBO Mol Med 6: 278–294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sandborn WJ, Bhandari BR, Fogel R, Onken J, Yen E, Zhao X et al (2016). Randomised clinical trial: a phase 1, dose‐ranging study of the anti‐matrix metalloproteinase‐9 monoclonal antibody GS‐5745 versus placebo for ulcerative colitis. Aliment Pharmacol Ther 44: 157–169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Santamaria S, Yamamoto K, Botkjaer K, Tape C, Dyson MR, McCafferty J et al (2015). Antibody‐based exosite inhibitors of ADAMTS‐5 (aggrecanase‐2). Biochem J 471: 391–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Santamaria S, Fedorov O, McCafferty J, Murphy G, Dudhia J, Nagase H et al (2017). Development of a monoclonal anti‐ADAMTS‐5 antibody that specifically blocks the interaction with LRP1. MAbs 9: 595–602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sato H, Takino T, Okada Y, Cao J, Shinagawa A, Yamamoto E et al (1994). A matrix metalloproteinase expressed on the surface of invasive tumour cells. Nature 370: 61–65. [DOI] [PubMed] [Google Scholar]
- Sela‐Passwell N, Kikkeri R, Dym O, Rozenberg H, Margalit R, Arad‐Yellin R et al (2011). Antibodies targeting the catalytic zinccomplex of activated matrix metalloproteinases show therapeutic potential. Nat Med 18: 143–147. [DOI] [PubMed] [Google Scholar]
- Shimanovich I, Mihai S, Oostingh GJ, Ilenchuk TT, Bröcker EB, Opdenakker G et al (2004). Granulocyte‐derived elastase and gelatinase B are required for dermal‐epidermal separation induced by autoantibodies from patients with epidermolysis bullosa acquisita and bullous pemphigoid. J Pathol 204: 519–527. [DOI] [PubMed] [Google Scholar]
- Shiraishi A, Mochizuki S, Miyakoshi A, Kojoh K, Okada Y (2015). Development of human neutralizing antibody to ADAMTS4 (aggrecanase‐1) and ADAMTS5 (aggrecanase‐2). Biochem Biophys Res Commun 469: 62–69. [DOI] [PubMed] [Google Scholar]
- Shiryaev SA, Remacle AG, Golubkov VS, Ingvarsen S, Porse A, Behrendt N et al (2013). A monoclonal antibody interferes with TIMP‐2 binding and incapacitates the MMP‐2‐activating function of multifunctional, pro‐tumorigenic MMP‐14/MT1‐MMP. Oncogene 2: e80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Song RH, Tortorella MD, Malfait AM, Alston JT, Yang Z, Arner EC et al (2007). Aggrecan degradation in human articular cartilage explants is mediated by both ADAMTS‐4 and ADAMTS‐5. Arthritis Rheum 56: 575–585. [DOI] [PubMed] [Google Scholar]
- Stanton H, Rogerson FM, East CJ, Golub SB, Lawlor KE, Meeker CT et al (2005). ADAMTS‐5 is the major aggrecanase in mouse cartilage in vivo and in vitro . Nature 434: 648–652. [DOI] [PubMed] [Google Scholar]
- Stocks M (2005). Intrabodies as drug discovery tools and therapeutics. Curr Opin Chem Biol 9: 359–365. [DOI] [PubMed] [Google Scholar]
- Suenaga N, Mori H, Itoh Y, Seiki M (2005). CD44 binding through the hemopexin‐like domain is critical for its shedding by membrane‐type 1 matrix metalloproteinase. Oncogene 24: 859–868. [DOI] [PubMed] [Google Scholar]
- Tallant C, García‐Castellanos R, Baumann U, Gomis‐Rüth FX (2010). On the relevance of the Met‐turn methionine in metzincins. J Biol Chem 285: 13951–13957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Talmi‐Frank D, Altboum Z, Solomonov I, Udi Y, Jaitin DA, Klepfish M et al (2016). Extracellular matrix proteolysis by MT1‐MMP contributes to influenza‐related tissue damage and mortality. Cell Host Microbe 20: 458–470. [DOI] [PubMed] [Google Scholar]
- Tam EM, Wu YI, Butler GS, Stack MS, Overall CM (2002). Collagen binding properties of the membrane type‐1 matrix metalloproteinase (MT1‐MMP) hemopexin C domain. The ectodomain of the 44‐kDa autocatalytic product of MT1‐MMP inhibits cell invasion by disrupting native type I collagen cleavage. J Biol Chem 277: 39005–39014. [DOI] [PubMed] [Google Scholar]
- Tape CJ, Willems SH, Dombernowsky SL, Stanley PL, Fogarasi M, Ouwehand W et al (2011). Cross‐domain inhibition of TACE ectodomain. Proc Natl Acad Sci U S A 108: 5578–5583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thomas MR, de Groot R, Scully MA, Crawley JT (2015). Pathogenicity of anti‐ADAMTS13 autoantibodies in acquired thrombotic thrombocytopenic purpura. EBioMedicine 2: 942–952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Udi Y, Grossman M, Solomonov I, Dym O, Rozenberg H, Moreno V et al (2015). Inhibition mechanism of membrane metalloprotease by an exosite‐swiveling conformational antibody. Structure 23: 104–115. [DOI] [PubMed] [Google Scholar]
- Visintin M, Caselli G, Chiusaroli R, Rovati LC (2012). Anti‐ADAMTS‐5 antibody, derivatives and uses thereof. WO2013153189A1.
- Woskowicz AM, Weaver SA, Shitomi Y, Ito N, Itoh Y (2013). MT‐LOOP‐dependent localization of membrane type I matrix metalloproteinase (MT1‐MMP) to the cell adhesion complexes promotes cancer cell invasion. J Biol Chem 288: 126–137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yamamoto K, Trad A, Baumgart A, Hüske L, Lorenzen I, Chalaris A et al (2012). A novel bispecific single‐chain antibody for ADAM17 and CD3 induces T‐cell‐mediated lysis of prostate cancer cells. Biochem J 445: 135–144. [DOI] [PubMed] [Google Scholar]
- Yamamoto K, Santamaria S, Botkjaer KA, Dudhia J, Troeberg L, Itoh Y et al (2017). Inhibition of shedding of low‐density lipoprotein receptor‐related protein 1 reverses cartilage matrix degradation in osteoarthritis. Arthritis Rheumatol 69: 1246–1256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang CY, Chanalaris A, Troeberg L (2017). ADAMTS and ADAM metalloproteinases in osteoarthritis – looking beyond the 'usual suspects. Osteoarthritis Cartilage 25: 1000–1009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ye J, Yuen SM, Murphy G, Xie R, Kwok HF (2017). Anti‐tumor effects of a ‘human & mouse cross‐reactive’ anti‐ADAM17 antibody in a pancreatic cancer model in vivo . Eur J Pharm Sci Eur J Pharm Sci 110: 62–69. [DOI] [PubMed] [Google Scholar]
- Zheng XL, Wu HM, Shang D, Falls E, Skipwith CG, Cataland SR et al (2010). Multiple domains of ADAMTS13 are targeted by autoantibodies against ADAMTS13 in patients with acquired idiopathic thrombotic thrombocytopenic purpura. Haematologica 95: 1555–1562. [DOI] [PMC free article] [PubMed] [Google Scholar]
