Cancer Letters 467 (2019) 50–57

Contents lists available at ScienceDirect

Cancer Letters

journal homepage: www.elsevier.com/locate/canlet

Mini-review ADAM : Emerging role and targeting of the non-catalytic domains T ∗ ∗ Nayanendu Saha , Dorothea Robev, Juha P. Himanen, Dimitar B. Nikolov

Sloan-Kettering Institute for Cancer Research, 1275 York Avenue, New York, NY, 10065, USA

ARTICLE INFO ABSTRACT

Keywords: ADAM proteases are multi domain transmembrane metalloproteases that cleave a range of surface ADAM10 and activate signaling pathways implicated in tumor progression, including those mediated by Notch, EFGR, and ADAM17 the Eph receptors. Consequently, they have emerged as key therapeutic targets in the efforts to inhibit tumor initiation and progression. To that end, two main approaches have been taken to develop ADAM antagonists: (i) Therapeutic antibody small molecule inhibitors, and (ii) monoclonal antibodies. In this mini-review we describe the distinct features of Notch signaling ADAM proteases, particularly of ADAM10 and ADAM17, their domain organization, conformational re- arrangements, regulation, as well as their emerging importance as therapeutic targets in cancer. Further, we highlight an anti-ADAM10 monoclonal antibody that we have recently developed, which has shown significant promise in inhibiting Notch signaling and deterring growth of solid tumors in pre-clinical settings.

1. Background substrate-specificities of the ADAMs are poorly understood [17]. They seem to be dictated, at least in part, by the ADAM D and C domains, Proteolytic processing and subsequent release of membrane-bound which mediate -protein, and specifically substrate-, in- proteins is a post-translational mechanism that governs the biological teractions. The D domains of ADAMs are also implicated in - activity of the cleaved molecules [1–3]. This process, referred to as mediated cell-adhesion. In the case of ADAM15, a consensus RGD se- ‘protein ectodomain shedding’, is mediated by membrane-anchored quence in the D domain [17] is needed for interaction with the α5β1 ADAM metalloproteases [4–9], which shed a wide range of membrane- and α5β3 integrins, while the importance of the C domain for proper attached proteins implicated in growth factor signaling, cell migration substrate cleavage has been documented for ADAM13 and ADAM17 and cell-adhesion [10]. In this mini-review, we will discuss the salient [8,18]. Swapping of the C domain of ADAM13 with that of ADAM10 features of this unique class of proteases, the regulation of their activity leads to loss of function and failure to induce hyperplasia in Xenopus via conformational rearrangements in their ectodomains and their po- laevis embryos [19], deterring major developmental events. Likewise, a tential as therapeutic targets during cancer progression. We will also chimeric construct of ADAM17, with its D + C domains replaced by discuss a novel anti-ADAM10 monoclonal antibody that has made sig- those of ADAM10, fails to shed interleukin (IL)-1R–II from the cell nificant headway towards treatment of solid tumors in pre-clinical surface of the fibroblast cell line EC-2 [18]. setting. ADAMs contain several conserved domains: an N-terminal signal 2. Conformational regulation of ADAMs and intracellular sequence followed by a pro-domain, a metalloprotease domain (MP), a signaling domain (D), a cysteine-rich domain (C), an EGF-like domain (except ADAM10 and ADAM17), a transmembrane domain and a cy- Cleavage by ADAM proteases is regulated at many levels [8,11 ]. It is toplasmic domain [11]. The signal sequence guides the inactive ADAM known that ADAM-mediated cleavage is essential for G-protein-coupled proenzymes into the secretory pathway [12,13]. The prodomain acts as [20,21] or stress-induced [22] EGF receptor transactivation, and it is a chaperone during trans-Golgi transport and also inhibits premature modulated by intracellular signaling in response to growth factors, MP domain activation [14,15]. The MP domain confers zinc-dependent phorbol esters, and changes in tyrosine kinase activity [8]. Interest- proteolytic activity and contains a consensus HEXGHXXGXXHD motif ingly, while ADAM family members harbor cytoplasmic protein- that coordinates the zinc atom [16]. Despite being well studied, the docking motifs [8], cytoplasmically-truncated ADAM10 and ADAM17

Abbreviations: MP, Metalloprotease domain; D, Disintegrin domain; C, Cysteine rich domain; VEGF, Vascular endothelial growth factor; OSCC, Oral squamous cell carcinoma; ScFv, Single chain variable fragment; PDI, Protein disulfide ; TNBC, Triple negative breast cancer; MMP, Matrix ∗ Corresponding authors. E-mail address: [email protected] (N. Saha). https://doi.org/10.1016/j.canlet.2019.10.003 Received 9 April 2019; Received in revised form 27 September 2019; Accepted 1 October 2019 0304-3835/ © 2019 Elsevier B.V. All rights reserved. N. Saha, et al. Cancer Letters 467 (2019) 50–57

progression [39,41,42]. The erbBs display de-regulated signaling in many human cancers due to overexpression and/or mutations, in- cluding in some of the most intractable and common tumors, with EGFR and HER2 prominent in breast, ovarian and colon cancer [43]. In ad- dition, elevated ADAM17-mediated cleavage of erbB2/erbB4 in tumors is associated with constitutive receptor activity and poor prognosis and undermines receptor-targeted therapies [44,45]. Eph receptors and their membrane-bound ephrin ligands control cell migration and positioning during normal and oncogenic develop- ment [46,47]. We documented that ADAM10 binds to Eph/ephrin re- ceptor/ligand complexes and cleaves the ephrin ligands in trans. This disrupts the Eph/ephrin molecular tethers between cells, causing de- adhesion and retraction [48], as well as termination of Eph signaling. This function of ADAM10 was found to be dependent on the Eph kinase activity [48]. ADAM10, thereby, acts as a switch between cell-cell ad- hesion and segregation in response to the Eph phosphorylation status. Fig. 1. ADAM domains and proposed conformations. The ‘closed’ and ‘open’ This is important in the context of Eph-dependent oncogenesis, where ectodomain conformations of ADAM10 and ADAM17 are proposed to reflect aberrant Eph expression can contribute to tumorigenesis [46,47]. fi alternate disul de bonding arrangements of the D + C region. Both ADAM10 Notch receptors (Notch1-4) are single pass transmembrane proteins and ADAM17 contain a CxxCC motif, a consensus site for PDI-mediated dis- that undergo regulated intramembrane proteolysis by ADAM10 and γ- ulphide exchange. Thus, their ectodomain conformations are influenced by secretase. The proteolytic steps are triggered by Notch binding to DLL/ redox effects. Jagged cell-surface-attached ligands. This initiates ADAM10-mediated shedding of the ligand [49] and receptor ectodomains [50]. Subse- are fully functional [23], suggesting only a minor role for the cyto- quently, release of the Notch intracellular domain (NICD) by γ-secretase plasmic domains in controlling downstream signaling. activates transcription of target , a pathway that is conserved in ADAM17 is suggested to be regulated by cysteine oxidation that all phyla [51]. Notch signaling controls cell fate and differentiation, modulates the conformation of its extracellular domain [24,25]. Fur- proliferation, epithelial-mesenchymal transition (EMT) and cell sur- thermore, the notion that the ADAM activity is regulated by the pre- vival, while de-regulated Notch signaling promotes progression of both “ ” “ ” “ ” sence of two distinct conformations active or open and latent or hematological and solid cancers [52]. Notch activity is particularly “ ” closed (Fig. 1) is supported by the crystal structures of related snake associated with a ‘stem-like’ phenotype in a subpopulation of tumor venom . Indeed, these structures reveal two radically cells termed as cancer stem cells (CSCs) or tumor initiating cells (TICs). ff fi di erent conformations resulting from alternative disul de-bonding The CSCs not only initiate and sustain tumor growth, but also promote arrangements of the D + C region [26,27]. metastasis, chemoresistance, and angiogenesis [53,54]. This is espe- fi Like other proteins undergoing protein disul de isomerase (PDI)- cially evident in colorectal cancer (CRC) [55], where endothelial-ex- fi mediated thiol disul de exchange, ADAM17 contains a conserved pressed Notch ligands promote paracrine signaling and stem cell phe- CxxCC motif, and mild reducing or oxidizing conditions alter its enzy- notype in tumor cells [56]. Accordingly, inhibition of Notch depletes fi matic activity [24]. Interestingly, facilitating disul de exchange via a CSCs and inhibits metastasis [57-60]. Notch signaling and CSCs are also thiol isomerase was reported to favor the inactive ADAM17 con- associated with drug resistance, and inhibition of Notch signaling is formation, while inhibiting the isomerase enhances the ADAM17 ac- widely reported to increase sensitivity to both chemo- and targeted tivity [25]. Other studies suggest that reactive oxygen species (ROS) therapies [61–65]. Importantly, it has been demonstrated that ADAM10 also regulate the activity of ADAM proteases through similar con- deletion is embryonic lethal in mice and the embryonic defects – formational changes in their ectodomains [28 33]. An important source match the defects observed in Notch deficient . This suggests of ROS at cell membranes is a family of NADPH oxidases (NOXs) that that the abnormalities associated with the absence of ADAM10 arise get stimulated by activated receptor tyrosine kinases (RTKs), including due to a failure to activate Notch signaling [57]. EGF, PDGF and receptors [34]. It has been elegantly demon- strated that an elevated level of ROS due to oxidative stress, common in 4. ADAM proteases as therapeutic targets tumors [35], stimulates ADAM-mediated shedding [30], cross-activa- tion of RTKs [29], and contributes to tumor resistance to therapy Since the ADAM proteases are involved in ectodomain shedding in a [36,37]. Furthermore, an elevated level of ROS inhibits protein tyrosine wide range of disease-related pathways, they are considered to be at- phosphatases (PTPs) by reversible oxidation of their conserved active- tractive targets for drug development. Studies with hepatocellular site cysteine residue [38], which in turn increases RTK activity. The carcinoma cell lines highlight the importance of ADAM10 as anti-tumor oxidation-dependent changes in the conformations of ADAM10 and therapeutic target, showing that downregulation of ADAM10 using ADAM17 suggest that RTK-stimulated ROS production could directly siRNA markedly suppresses cell proliferation, cell migration and cell activate ADAMs. This implies a mechanism where conformational invasion in vitro, as well as tumor growth in vivo [66]. In addition, changes in the ADAM ectodomains mediate ADAM-RTK cross activa- ADAM10 is over-expressed in oral squamous cell carcinoma (OSCC), tion. where it contributes to OSCC invasion by interacting with integrin αvβ6 [67]. Likewise, ADAM17 is over-expressed in esophageal squamous cell 3. Oncogenic substrates of ADAM10 and ADAM17 carcinoma [68] and plays a significant role in the progression of glioma by upregulating the EGFR-P13K-AKT pathway [69]. Furthermore, mi- ADAM10 and ADAM17 shed various cell-surface proteins and acti- croarray analysis revealed that ADAM17 is over-expressed on the sur- vate signaling pathways important in oncogenic development. face of several types of cancer cells, including ovarian, breast and Prominent oncogenic substrates include receptors/ligands of the erbB, prostate [70]. In ovarian cancer, ADAM10 and ADAM17 cleave the Eph and Notch families, cytokines, cadherins, FAS ligand, Slit and L- calcium independent molecule Nectin-4 [71]. The cleaved selectin [39]. ErbB ligands are initially cell-surface tethered and their ectodomain of Nectin-4 facilitates formation of spheroids that even- proteolytic release by ADAM10 and ADAM17 [40 ] is a key regulatory tually invade the mesothelial layer to establish tumors at secondary switch to trigger downstream autocrine signaling that drives tumor sites [72].

51 N. Saha, et al. Cancer Letters 467 (2019) 50–57

ADAM9, another member of the family, is overexpressed in several documents the various ADAM proteases implicated in major cancer cancers, including ovarian, clear cell carcinoma [73], lung adeno- types. carcinoma [74], glioma, gastric, breast, and liver cancers [75]. In liver Owing to the importance and therapeutic potential of ADAM pro- cancer, ADAM9 was shown to evade tumor immunity by cleaving the teases in cancer progression, attempts have been made to develop MHC class 1 polypeptide–related sequence A. Interestingly, it was es- ADAM inhibitors to impede cancer growth. The two major approaches tablished that ADAM9 interacts with β1 integrins (but does not cleave taken so far are discussed in the following paragraphs. them) promoting melanoma, thus highlighting the importance of non- proteolytic ADAM functions in cancer progression [76]. Likewise, the 5. Small molecule ADAM inhibitors cysteine-rich domain of ADAM12 is necessary for tumor cell adhesion [77]. The expression levels of ADAM12 were found to be significantly Most of the small molecule ADAM inhibitors described to date are elevated in small cell lung cancer patients [78]. Amongst other cancer hydroxamate-based and are either specific for ADAM17 or selective for types, ADAM12 is responsible for cavernous sinus invasion in pituitary both ADAM10 and ADAM17. These inhibitors chelate the zinc from the adenomas [79]. Owing to the low expression levels of ADAM12 in of the proteases, thereby rendering them inactive. Amongst normal tissues, as compared to its high expression levels in several the dual inhibitors for ADAM10 and ADAM17, INCB3619 was one of cancers (including both mRNA and protein), ADAM12 can be a useful the first to be investigated for anti-cancer activity [42,93,94]. In in vitro biomarker [80]. ADAM15 mRNA and protein levels are increased in assays, the compound reduces the shedding of an array of EGFR ligands, prostate cancer, where it triggers angiogenesis and angioinvasion of which increases the susceptibility of non-small cell lung cancer cells to tumor cells causing unrestrained tumor growth [81]. In ovarian cancer, the EGFR tyrosine kinase inhibitor gefitinib [42]. Unlike the first-gen- ADAM15 disrupts the binding of αvβ3 integrin to its extracellular eration inhibitors, INCB3619 is remarkably free of musculoskeletal matrix ligand , thereby regulating tumor migration and in- toxicity in models. INCB3619 also inhibits non-small cell lung, vasion. Mechanistically, the RGD-binding motif of ADAM15 interacts breast, head and neck carcinomas in animal models [42,93,94]. Of late, with αvβ3 integrin and loosens the adhesion of tumor cells to the un- another structurally related inhibitor, INCB7839, underwent early derlying matrix [82]. ADAM8 promotes growth and progression of clinical trials in HER-2 positive breast cancer patients with no evidence triple negative breast cancer (TNBC) that lacks estrogen, progesterone of musculoskeletal side effects or bone marrow toxicity. This particular and HER-2 receptor gene expression. In pancreatic cancer cells, ADAM8 inhibitor decreases the shedding of EGFR and HER-2 ligands [95]. Be- enhances the binding of β1 integrin to endothelial cells. Thus, it pro- sides the INC class of dual inhibitors, the PF-series, including PF- motes tumor cell migration and invasion by augmenting the ERK1/2 5480090, was found to be specific for ADAM17 and does not inhibit signaling pathway [83,84]. Recently, ADAM23, was found to be in- ADAM10 [96]. PF-5480090 inhibits proliferation of a wide panel of volved in breast cancer progression and dissemination of circulating breast cancer cell lines independent of their molecular subtype [97]. tumor cells [85]. ADAM28 promotes lung metastasis by cleaving and Specifically, in the case of TNBC where ADAM17 is expressed at higher inactivating the von Willebrand factor, VWF. VWF is a proapoptotic level than in non-TNBC, PF-5480900 significantly decreases (i) the re- protein and its inactivation prolongs cancer cell survival and metastasis lease of the EGFR ligand TGF alpha and (ii) the levels of phosphorylated [86,87]. In TNBC, ADAM33 has been reported to be downregulated by EGFR, leading to proliferation inhibition of TNBC [97]. In addition to DNA hypermethylation, suggesting that this could be poten- small molecule compounds, it has been shown that small hairpin RNAs, tially used as a biomarker for TNBC and other basal-like breast cancers specific to ADAM17, significantly inhibit cell proliferation, invasion [88]. and tumor growth of non-small cell lung cancer in vitro [98]. Interestingly, some of the catalytically ‘dead’ human ADAM pro- Little progress has been made with regard to ADAM10-specific small teases, such as ADAM11 and ADAM29, were shown to promote integrin molecule inhibitors. One such molecule, GI254023X, blocks migration α4-dependent adhesion of immune cells. The studies revealed that the and invasion of breast cancer cell lines with hardly any effect on pro- disintegrin domains of these ADAM proteases mediate cell-cell adhe- liferation. In cell-based assays, GI254023X impedes shedding of inter- sions via interactions with the integrins α4β1 and α4β7[89]. leukin receptors and chemokine ligands [99–101]. Amongst the other The ADAMTS (A Disintegrin and Metalloprotease domains with ADAM proteases, a six-amino acid cyclic peptide, designated as BK- thrombospondin motifs) are secreted, multi-domain endopeptidases, 1361, inhibits the activation of ADAM8 and decreases tumor burden and close relatives of the ADAMs [90]. They have been implicated in and metastasis of implanted pancreatic tumor cells. Specifically, it both oncogenic and tumor-suppressive signaling events. Specifically, downregulates the ERK1/2 signaling pathway [83,84]. No small mo- they regulate the inflammatory microenvironment of tumors. For in- lecule inhibitors have been reported for ADAM28, though in animal stance, ADAMTS4 causes macrophage infiltration in the environs of models of lung cancer, ADAM28-targeting siRNA inhibits metastasis colorectal cancer [91]. Elevated expression of ADAMTS 1, 8, 9 and 18 [86]. ADAM15 is known to be functionally associated with the pro- were also observed in pancreatic cancer patient samples [92]. Table 1 gression of bladder cancer. Indeed, knock-down of ADAM15 in a human xenograft model of bladder cancer causes 45% tumor regression com- pared to controls [102]. A novel ADAM15-specific sulfonamide in- Table 1 hibitor, designed by structural modeling of the catalytic domain, also ADAM proteases and associated major cancer types. decreases tumor growth in animal model of bladder cancer [102]. ADAM/ADAMTS Cancer types References 6. Monoclonal antibodies targeting the ADAMs ADAM8 Triple negative breast cancer, Pancreatic [83,84] ADAM9 Ovarian, lung, glioma, breast, gastric, liver [73–76] ADAM12 Small cell lung, pituitary adenomas [77–80] The second approach for therapeutic intervention entails the gen- ADAM15 Ovarian [81,82] eration of monoclonal antibodies (mAbs) targeting the ADAMs. ADAM10 Colon, hepatocellular, oral squamous cell [66,67,114] Compared to small molecule inhibitors, mAbs can be highly target- carcinoma fi ff ADAM17 Esophageal, glioma, ovarian, breast, [68–71] speci c, thereby reducing the risk of toxic side e ects. Of late, a mAb prostate that binds to both the catalytic (MP) and disintegrin-cysteine rich do- ADAM23 Breast [85] mains of ADAM17 has been shown to have significant therapeutic po- ADAM28 Lung [86,87] tential [103,104]. The mAb D1 (A12) inhibits tumor growth in an an- ADAM33 Breast [88] imal model of ovarian cancer and blocks the shedding of ADAM17 ADAMTS4 Colorectal [91] ADAMTS 1, 8, 9, 18 Pancreatic [92] substrates, including TNF-alpha, TGF-alpha, HG-EGF and amphiregulin [103–105]. In a mouse model, the mAb is highly stable in circulation

52 N. Saha, et al. Cancer Letters 467 (2019) 50–57 and retains its ability to bind ADAM17 over a prolonged period of time MP + D + C domain architectures [26,27]. This comparison (Fig. 3A) [103]. Another anti-ADAM17 mAb, MEDI3622, inhibits cancer pro- shows that the MP domain, residing within the concave side of the gression in several different pre-clinical models [106]. Mutagenesis and D + C region, occupies the same space where the 8C7 mAb binds. It is, molecular docking studies revealed that this mAb recognizes a unique therefore, likely that 8C7 displaces the ADAM10 MP domain upon surface loop present in the MP domain of ADAM17 [107]. In cell–based binding, thus inhibiting substrate cleavage on the cell surface. The re- studies, MEDI3622 blocks the release of known ADAM17 substrates, cent crystal structure of the complete ADAM10 ectodomain corrobo- including amphiregulin, epiregulin and heregulin [106]. rates this hypothesis (Fig. 3B). In that structure, ADAM10 adopts a In another study, a mAb (A300E) recognizing the membrane-prox- closed conformation, with the MP and the D + C domains positioned imal cysteine-rich region of human ADAM17 was reported. It was fur- similarly to what was observed in the Atragin structure [115]. ther modified to generate a recombinant scFv (single-chain variable fragment) containing the variable regions of the heavy and light chains. 7.1. Future prospects Using this scFv, a bispecific antibody was constructed that recognizes ADAM17 on tumor cells and CD3 on T-cells. In the presence of primary Targeting the MP domains of ADAM proteases, particularly of peripheral blood mononuclear cells, this bispecific antibody specifically ADAM10 and ADAM17, for the treatment of various diseases, has been kills prostate tumor cells expressing ADAM17 on the cell surface [108]. unsuccessful so far mostly because the proteinase active site is highly A300E, when coupled to doxorubicin or Pseudomonas exotoxin A, is conserved, resulting in toxicity due to the interactions of drugs with able to kill the MDA-MB-231 TNBC cell line in an ADAM17-dependent other matrix metalloproteinases. Recently, it has been shown that the manner [109]. Amongst the other ADAM proteases, a monoclonal an- non-catalytic ADAM10 and ADAM17 domains, specifically the D and C tibody raised against the catalytic site of ADAM9 suppresses cancer cell domains, confer substrate-specificities, and it is expected that future proliferation and exhibits anti-tumor activity in a gastric cancer xeno- efforts will concentrate on targeting these domains for successful graft model [110]. ADAM8 was found to promote breast cancer de- therapeutic intervention. This endeavor could also be extrapolated to velopment and brain metastasis in mouse models, and a commercial other ADAM family members, which differ from ADAM10 and ADAM17 antibody to ADAM8 blocks transendothelial migration of breast cancer in that they contain an additional EGF domain. For instance, ADAM9, cells in in vitro assays [111]. ADAM12, ADAM13 and ADAM15 [19,75,77,82] bind to a variety of integrins via their non-catalytic domains promoting adhesion of tumor 7. Mechanism of action and therapeutic potential of the 8C7 anti- cells. Likewise, the ancillary disintegrin, cysteine-rich and thrombos- ADAM10 mAb pondin domains of ADAMTS dictate substrate-specificity, and thus regulate the diverse functions of these secreted proteases [90]. Struc- Though ADAM10 is implicated in several different cancers, in- tural studies have already shown specific molecular regions and sur- cluding breast, pancreatic, melanoma and bladder carcinoma, no faces outside of the ADAM MP domain that mediate ADAM-substrate ADAM10 specific mAb has been developed so far for therapeutic in- interactions, providing attractive targets for novel small-molecule and tervention. Furthermore, Notch is the most prominent tumor-related antibody therapeutics. Furthermore, a large and ever-growing amount ADAM10 target, and previous attempts to inhibit tumorigenic Notch of DNA sequencing data from healthy individuals and patients provides signaling by blocking the γ-secretase activity or disrupting the forma- a great opportunity to not only understand better the molecular me- tion of Notch-ligand complexes caused intestinal toxicity [112]. chanisms of ADAM-mediated signaling, but to also identify and/or va- Nevertheless, Notch-inhibitory mAbs that do not exhibit toxicity have lidate therapeutic target regions on the molecular surface of the ADAM proved to be promising cancer therapeutic leads [113]. We, therefore, family members. One such source is the sequencing data from the generated a highly specific anti-ADAM10 mAb (8C7) for the potential Center for Molecular Oncology, Memorial Sloan Kettering Cancer treatment of solid tumors. This antibody binds to the cysteine-rich Center [disseminated through cBioportal, http://www.cbioportal.org/ domain of ADAM10 and targets not only the rapidly dividing cells at the [116,117]]. The use of genomic and proteomic data should also provide invasive font of the tumors, but also the undifferentiated tumor stem in the future the basis for “personalized/precision medicine” as well as cells. We documented that 8C7 mAb primarily acts via inhibiting Notch novel “targeted therapeutics” developments [118,119]. signaling in tumors. It also effectively prevents tumor relapse after chemotherapy in pre-clinical models without exhibiting any dose-lim- iting toxicity in animals [114]. Conflicts of interest Our in vitro and in vivo data indicate that 8C7 recognizes an active conformation of ADAM10 prevalent in tumors, as opposed to the in- The authors declare that they have no competing interest. active/latent ADAM10 ectodomain conformation present in normal tissues. It should be noted that currently there is no structural in- CRediT authorship contribution statement formation regarding the open, activated ADAM10 conformation, and only the structure of the closed, autoinhibited conformation has been Nayanendu Saha: Conceptualization, Writing - review & editing, reported, where access to the proteinase active site is blocked [115]. Writing - original draft. Dorothea Robev: Software, Writing - review & The transition to the active ADAM conformation, as discussed in the editing. Juha P. Himanen: Writing - review & editing, Formal analysis, previous sections of this review, is dependent on disulfide isomerization Funding acquisition. Dimitar B. Nikolov: Formal analysis, Writing - and oxidative conditions of the tumor microenvironment [29]. Conse- review & editing, Funding acquisition. quently, in tumor cells, oxidizing conditions promote binding of the 8C7 mAb to ADAM10, while reducing conditions inhibit binding. The crystal structure of 8C7 bound to the ADAM10 D + C domain Acknowledgements and Funding [114] revealed the ADAM10 epitope recognized by 8C7. This ADAM10 region is stabilized by two intra-molecular disulfide bonds, C594–C639 Our studies are supported by National Institutes of Health and C632–C645, which are part of a CxxCC motif, a consensus PDI [R21CA185930 and R01NS038486 to D.B.N and 5RO1NS096956-03/ target sequence (Fig. 2). Mutating the CxxCC motif to AxxAC abrogates RES510708 to JPH]; The Experimental Therapeutics Center of binding of 8C7 to ADAM10. Memorial Sloan-Kettering, support from Mr. William H. and Mrs. Alice To understand how the 8C7 mAb inhibits ADAM10 function, the Goodwin and the Commonwealth Foundation for Cancer Research ADAM10-8C7 complex structure was compared to the structures of full- [2013–2015 and 2018–2020 to D.B.N]; Functional Genomics Initiative length snake venom metalloproteases VAP1 and Atragin, with similar [2015–2017 and 2019–2020 to D.B.N.]

53 N. Saha, et al. Cancer Letters 467 (2019) 50–57

Fig. 2. A close up-view of the disulfide- bonding pattern in the mAb (8C7)-bound ADAM10. The disulfide forming cysteines are shown as blue and yellow spheres. The ADAM10 D and C domains are represented in red. The heavy chain of the 8C7 mAb is in green and the light chain in wheat [114]. (For interpretation of the references to color in this figure legend, the reader is referred to the Web version of this article.)

Fig. 3. Comparison of the mAb 8C7-bound ADAM10 D + C structure with the structures of the related snake venom metalloproteinases Atragin and VAP1 (3A), as well as the structure of the complete ADAM10 ectodomain (3B)[115].

54 N. Saha, et al. Cancer Letters 467 (2019) 50–57

References axon repellent, Science 289 (2000) 1360–1365. [32] T. Rzymski, A. Petry, D. Kracun, F. Reisz, L. Pike, A.L. Harris, A. Gorlach, The unfolded protein response controls induction and activation of ADAM17/TACE by [1] N.M. Hooper, E.H. Karran, A.J. Turner, Membrane protein secretases, Biochem. J. severe hypoxia and ER stress, Oncogene 31 (31) (2012) 3621–3634. – 321 (Pt 2) (1997) 265 279. [33] B. Wang, T. Mysliwiec, S.M. Feller, B. Knudsen, H. Hanafusa, G.D. Kruh, Proline- [2] N.M. Hooper, A.J. Turner, Protein processing mechanisms: from angiotensin- rich sequences mediate the interaction of the Arg protein tyrosine kinase with Crk, converting enzyme to Alzheimer's disease, Biochem. Soc. Trans. 28 (2000) Oncogene 13 (1996) 1379–1385. – 441 446. [34] P. Chiarugi, P. Cirri, Redox regulation of protein tyrosine phosphatases during [3] J. Massague, A. Pandiella, Membrane-anchored growth factors, Annu. Rev. receptor tyrosine kinase signal transduction, Trends Biochem. Sci. 28 (2003) – Biochem. 62 (1993) 515 541. 509–514. [4] J.D. Becherer, C.P. Blobel, Biochemical properties and functions of membrane- [35] C.C. Benz, C. Yau, Ageing, oxidative stress and cancer: paradigms in parallax, Nat. anchored metalloprotease-disintegrin proteins (ADAMs), Curr. Top. Dev. Biol. 54 Rev. Cancer 8 (2008) 875–879. – (2003) 101 123. [36] M.V. Karamouzis, P.A. Konstantinopoulos, A.G. Papavassiliou, Targeting MET as a [5] R.A. Black, J.M. White ADAMs, Focus on the protease domain, Curr. Opin. Cell strategy to overcome crosstalk-related resistance to EGFR inhibitors, Lancet Oncol. – Biol. 10 (1998) 654 659. 10 (2009) 709–717. [6] F. Kheradmand, Z. Werb, Shedding light on : role in growth and de- [37] V. Pillay, L. Allaf, A.L. Wilding, J.F. Donoghue, N.W. Court, S.A. Greenall, – velopment, Bioessays: News Rev. Mol. Cell. Develop. Biol. 24 (2002) 8 12. A.M. Scott, T.G. Johns, The plasticity of oncogene addiction: implications for fi [7] M.L. Moss, J.W. Bartsch, Therapeutic bene ts from targeting of ADAM family targeted therapies directed to receptor tyrosine kinases, Neoplasia 11 (2009) – members, Biochemistry 43 (2004) 7227 7235. 448–458. ff [8] P. Primako , D.G. Myles, The ADAM gene family: surface proteins with adhesion [38] B.G. Neel, H. Gu, L. Pao, The 'Shp'ing news: SH2 domain-containing tyrosine – and protease activity, Trends Genet. 16 (2000) 83 87. phosphatases in cell signaling, Trends Biochem. Sci. 28 (2003) 284–293. ff [9] J. Schlondor , C.P. Blobel, Metalloprotease-: modular proteins capable [39] G. Murphy, The ADAMS: signalling scissors in the tumour microenvironment, Nat. of promoting cell-cell interactions and triggering signals by protein-ectodomain Rev. Cancer 8 (2008) 929–941. – shedding, J. Cell Sci. 2 (Pt 21) (1999) 3603 3617. [40] R.C. Harris, E. Chung, R.J. Coffey, EGF receptor ligands, Exp. Cell Res. 284 (1) ff [10] T. Klein, R. Bischo , Active metalloproteases of the A Disintegrin and (2003) 2–13. Metalloprotease (ADAM) family: biological function and structure, J. Proteome [41] C.P. Blobel, G. Carpenter, M. Freeman, The role of protease activity in ErbB – Res. 10 (2011) 17 33. biology, Exp. Cell Res. 315 (4) (2009) 671–682. [11] C.P. Blobel ADAMs, Key components in EGFR signalling and development, Nat. [42] B.-B.S. Zhou, M. Petyon, B. He, C. Liu, L. Girard, E. Caudler, Y. Lo, F. Baribaud, – Rev. Mol. Cell Biol. 6 (2005) 32 43. I. Mikami, N. Reguart, et al., Targeting ADAM-mediated ligand cleavage to inhibit [12] A. Anders, S. Gilbert, W. Garten, R. Postina, F. Fahrenholz, Regulation of the HER2 and EGFR pathways in non-small cell lung cancer, Cancer Cell 10 (1) (2006) alpha-secretase ADAM10 by its prodomain and proprotein convertases, FASEB 39–50. ff (Fed. Am. Soc. Exp. Biol.) J.: O . Publ. Fed. Am. Soc. Exper. Biol. 15 (2001) [43] Y. Yarden, M.X. Sliwkowski, Untangling the ErbB signaling network, Nat. Rev. – 1837 1839. Mol. Cell Biol. 2 (2001) 127–137. [13] F. Loechel, M.T. Overgaard, C. Oxvig, R. Albrechtsen, U.M. Wewer, Regulation of [44] C. Rio, J.D. Buxbaum, J.J. Peschon, G. Corfas, alpha-con- human ADAM 12 protease by the prodomain. Evidence for a functional cysteine verting enzyme is required for cleavage of erbB4/HER4, J. Biol. Chem. 275 (14) – switch, J. Biol. Chem. 274 (1999) 13427 13433. (2000) 10379–10387. [14] F. Fahrenholz, S. Gilbert, E. Kojro, S. Lammich, R. Postina, Alpha-secretase activity [45] S.E. Wang, B. Xiang, M. Guix, M.G. Olivares, J. Parker, C.H. Chung, A. Pandiella, fl of the disintegrin metalloprotease ADAM 10. In uences of domain structure, Ann. C.L. Artiega, Transforming growth factor beta engaged TACE and ErbB3 to acti- – N. Y. Acad. Sci. 920 (2000) 215 222. vate phosphatidylinositol-3 kinase/Akt in ErbB2-overexpressing breast cancer and [15] D.F. Seals, S.A. Courtneidge, The ADAMs family of metalloproteases: multidomain desensitizes cells to trastuzumab, Mol. Cell. Biol. 28 (18) (2008) 5605–5620. – proteins with multiple functions, Genes Dev. 17 (2003) 7 30. [46] E. Nievergall, M. Lackmann, P.W. Janes, Eph-dependent cell-cell adhesion and [16] T.G. Wolfsberg, J.M. White, ADAM Metalloproteinases, Handbook of Proteolytic segregation in development and cancer, Cell. Mol. Life Sci. 69 (2012) 1813-184. – , Academic Press, London, 1998, pp. 709 714. [47] E.B. Pasquale, Eph receptors and ephrins in cancer: bidirectional signaling and [17] J.M. White ADAMs, Modulators of cell-cell and cell-matrix interactions, Curr. beyond, Nat. Rev. Cancer 10 (2010) 165–180. – Opin. Cell Biol. 15 (2003) 598 606. [48] P.W. Janes, N. Saha, W.A. Barton, M.V. Kolev, S.H. Wimmer-Kleikamp, [18] P. Reddy, J.L. Slack, R. Davis, D.P. Cerretti, C.J. Kozlosky, R.A. Blanton, D. Shows, C.P. Blobel, J.P. Himanen, M. Lackmann, D.B. Nikolov, Adam meets Eph: an J.J. Peschon, R.A. Black, Functional analysis of the domain structure of tumor ADAM substrate recognition module acts as a molecular switch for ephrin cleavage – necrosis factor-alpha converting enzyme, J. Biol. Chem. 275 (2000) 14608 14614. in trans, Cell 123 (2) (2005) 291–304. [19] K.M. Smith, A. Gaultier, H. Cousin, D. Alfandari, J.M. White, D.W. DeSimone, The [49] M.J. LaVoie, D.J. Selkoe, The notch ligands, jagged and delta, are sequentially cysteine-rich domain regulates ADAM protease function in vivo, J. Cell Biol. 159 processed by α-secretase and presenilin/γ-secretase and release signaling frag- – (5) (2002) 893 902. ments, J. Biol. Chem. 278 (2003) 34427–34437. [20] N. Prenzel, E. Zwick, H. Daub, M. Leserer, R. Abraham, C. Wallasch, A. Ullrich, [50] R. Kopan, M.X.G. Ilagan, The canonical notch signaling pathway: unfolding the EGF receptor transactivation by G-protein-coupled receptors requires metallo- activation mechanism, Cell 137 (2009) 216–233. – proteinase cleavage of proHB-EGF, Nature 402 (1999) 884 888. [51] S.J. Bray, Notch signalling: a simple pathway becomes complex, Nat. Rev. Mol. [21] Y. Yan, K. Shirakabe, Z. Werb, The metalloprotease Kuzbanian (ADAM10) med- Cell Biol. 7 (2006) 678–689. iates the transactivation of EGF receptor by G protein-coupled receptors, J. Cell [52] P. Ranganathan, K.L. Weaver, A.J. Capobianco, Notch signalling in solid tumours: – Biol. 158 (2002) 221 226. a little bit of everything but not all the time, Nat. Rev. Cancer 11 (2011) 338–351. [22] O.M. Fischer, S. Hart, A. Gschwind, A. Ullrich, EGFR signal transactivation in [53] I. Espinoza, R. Pochampally, F. Xing, K. Watabe, L. Miele, Notch signaling: tar- – cancer cells, Biochem. Soc. Trans. 31 (2003) 1203 1208. geting cancer stem cells and epithelial-to-mesenchymal transition, OncoTargets [23] P.W. Janes, S.H. Wimmer-Kleikamp, A.S. Frangakis, K. Treble, B. Griesshaber, Ther. 6 (2013) 1249–1259. O. Sabet, M. Grabenbauer, A.Y. Ting, P. Saftig, P.I. Bastianens, M. Lackmann, [54] G. Fillipo, Giancotti. Mechanisms governing metastatic dormancy and reactiva- Cytoplasmic relaxation of active Eph controls ephrin shedding by ADAM10, PLoS tion, Cell 155 (2013) 750–764. Biol. 7 (2009) e1000215. [55] S. Miyamoto, D.W. Rosenberg, Role of Notch signaling in colon homeostasis and [24] Y. Wang, A.H. Herrera, Y. Li, K.K. Belani, B. Walcheck, Regulation of mature carcinogenesis, Cancer Sci. 102 (2011) 1938–1942. ADAM17 by redox agents for L-selectin shedding, J. Immunol. 182 (2009) [56] J. Lu, X. Ye, F. Fan, L. Xia, R. Bhattacharya, S. Bellister, F. Tozzi, E. Sceusi, – 2449 2457. Y. Zhou, I. Tachibana, D.M. Maru, D.H. Hawke, J. Rak, S.A. Mani, P. Zweidler- [25] S.H. Willems, C.J. Tape, P.L. Stanley, N.A. Taylor, I.G. Mills, D.E. Neal, McKay, L.M. Ellis, Endothelial cells promote the colorectal cancer stem cell phe- ff J. McCa erty, G. Murphy, Thiol negatively regulate the cellular notype through a soluble form of jagged-1, Cancer Cell 23 (2013) 171–185. – shedding activity of ADAM17, Biochem. J. 428 (2010) 439 450. [57] D. Hartmann, B. de Strooper, L. Serneels, K. Craessaerts, A. Herreman, W. Annaert, [26] H.H. Guan, K.S. Goh, F. Davamani, P.L. Wu, Y.W. Huang, J. Jeyakanthan, L. Umans, T. Lübke, A. Lena Illert, K. von Figura, P. Saftig, The disintegrin/me- W.G. Wu, C.J. Chen, Structures of two elapid snake venom metalloproteases with talloprotease ADAM 10 is essential for Notch signalling but not for alpha-secretase fi distinct activities highlight the disul de patterns in the D domain of activity in fibroblasts, Hum. Mol. Genet. 11 (21) (2002) 2615–2624. – family proteins, J. Struct. Biol. 169 (3) (2010) 294 303. [58] X. Fan, L. Khaki, T.S. Zhu, M.E. Soules, C.E. Talsma, N. Gul, C. Koh, J. Zhang, [27] S. Takeda, T. Igarashi, H. Mori, S. Araki, Crystal structures of VAP1 reveal ADAMs' Y.M. Li, J. Maciaczyk, G. Nikkhah, F. Dimeco, S. Piccirillo, A.L. Vescovi, ff MDC domain architecture and its unique C-shaped sca old, EMBO J. 25 (2006) C.G. Eberhart, NOTCH pathway blockade depletes CD133-positive glioblastoma – 2388 2396. cells and inhibits growth of tumor neurospheres and xenografts, Stem Cells 28 [28] L. Atapattu, M. Lackmann, P.W. Janes, The role of proteases in regulating Eph/ (2010) 5–16. – ephrin signaling, Cell Adhes. Migrat. 8 (2014) 294 307. [59] P.M. McGowan, M. Mullooly, F. Caiazza, S. Sukor, S.F. Madden, A.A. Maguire, [29] O.M. Fischer, S. Giordano, P.M. Comoglio, A. Ullrich, Reactive oxygen species A. Pierce, E.W. McDermott, J. Crown, N. O'Donovan, M.J. Duffy, ADAM-17: a mediate Met receptor transactivation by G protein-coupled receptors and the novel therapeutic target for triple negative breast cancer, Ann. Oncol. 24 (2013) epidermal growth factor receptor in human carcinoma cells, J. Biol. Chem. 279 362–369. – (2004) 28970 28978. [60] J. Domingo-Domenech, S.J. Vidal, V. Rodriguez-Bravo, M. Castillo-Martin, [30] O.M. Fischer, S. Hart, A. Gschwind, N. Prenzel, A. Ullrich, Oxidative and osmotic S.A. Quinn, R. Rodriguez-Barrueco, D.M. Bonal, E. Charytonowicz, N. Gladoun, stress signaling in tumor cells is mediated by ADAM proteases and heparin-binding J. de la Iglesia-Vicente, D.P. Petrylak, M.C. Benson, J.M. Silva, C. Cordon-Cardo, – epidermal growth factor, Mol. Cell. Biol. 24 (2004) 5172 5183. Suppression of acquired docetaxel resistance in prostate cancer through depletion fi [31] M. Hattori, M. Oster eld, J.G. Flanagan, Regulated cleavage of a contact-mediated of notch- and hedgehog-dependent tumor-initiating cells, Cancer Cell 22 (2012)

55 N. Saha, et al. Cancer Letters 467 (2019) 50–57

373–388. U. Schlomann, M. Parsons, J.W. Bartsch, G.E. Sonenshein, ADAM8 as a drug target [61] M. Fischer, W.C. Yen, A.M. Kapoun, M. Wang, G. O'Young, J. lewicki, A. Gurney, in pancreatic cancer, Nat. Commun. 28 (2015) 6175. T. Hoey, Anti-DLL4 inhibits growth and reduces tumor-initiating cell frequency in [85] I. Zmetakova, L. Kalinkova, B. Smolkova, V. Horvathova, Z. Cierna, L. Danihel, colorectal tumors with oncogenic KRAS mutations, Cancer Res. 71 (2011) M. Bohac, T. Sedlackova, G. Minarik, M. Karaba, J. Benca, M. Cihova, 1520–1525. V. Buocikova, S. Miklikova, M. Mego, I. Fridrichova, A disintegrin and metallo- [62] J. Li, R.C. Sainson, C.E. Oon, H. Turley, R. Leek, H. Sheldon, E. Bridges, W. Shi, protease 23 hypermethylation predicts decreased disease-free survival in low-risk C. Snell, E.T. Bowden, H. Wu, P.S. Chowdhury, A.J. Russell, C.P. Montgomery, breast cancer patients, Cancer Sci. 110 (5) (2019) 1695–1704. R. Poulsom, H.L. Harris, DLL4-Notch signaling mediates tumor resistance to anti- [86] A.M. Fourie, F. Coles, V. Moreno, L. Karlsson, Catalytic activity of ADAM8, VEGF therapy in vivo, Cancer Res. 71 (2011) 6073–6083. ADAM15, and MDC-L (ADAM28) on synthetic peptide substrates and in ectodo- [63] S.M. McAuliffe, S.L. Morgan, G.A. Wyant, L.T. Tran, K.W. Muto, Y.S. Chen, main cleavage of CD23, J. Biol. Chem. 278 (33) (2003) 30469–30477. K.T. Chin, J.C. Partridge, B.B. Poole, K.H. Cheng, J. Daggett, K. Cullen, E. Kantoff, [87] Y. Mitsui, S. Mochizuki, T. Kodama, M. Shimoda, T. Ohtsuka, T. Shiomi, K. Hasselbatt, J. Berkowitz, M.G. Muto, R.S. Berkowitz, J.C. Aster, U.A. Matulonis, M. Chijiiwa, T. Ikeda, M. Kitajima, Y. Okada, ADAM28 is overexpressed in human D.M. Dinulescu, Targeting Notch, a key pathway for ovarian cancer stem cells, breast carcinomas: implications for carcinoma cell proliferation through cleavage sensitizes tumors to platinum therapy, Proc. Natl. Acad. Sci. 109 (2012) of insulin-like growth factor binding protein-3, Cancer Res. 66 (20) (2006) E2939–E2948. 9913–9920. [64] R.D. Meng, C.C. Shelton, Y.M. Li, L.X. Qin, D. Notterman, P.B. Paty, Gamma- [88] G.C. Manica, C.F. Ribeiro, M.A. Oliveira, I.T. Pereira, A. Chequin, E.A. Ramos, Secretase inhibitors abrogate oxaliplatin-induced activation of the Notch-1 sig- L.M. Klassen, S.M. Zanata, L. Noronha, I. Rabinovich, F.F. Costa, E.M. Souza, naling pathway in colon cancer cells resulting in enhanced chemosensitivity, G. Klassen, Down regulation of ADAM33 as a predictive biomarker of aggressive Cancer Res. 69 (2009) 573–582. breast cancer, Sci. Rep. 7 (2017) 44414. [65] C.R. Timme, M. Gruidi, T.J. Yeatman, Gamma-secretase inhibition attenuates [89] L. Wang, J.A. Hoggard, E.D. Korleski, G.V. Long, B.C. Ree, K. Hensley, S.R. Bond, oxaliplatin-induced apoptosis through increased Mcl-1 and/or Bcl-xL in human T.G. Wolfsberg, J. Chen, T. Zeczycki, L.C. Bridges, Multiple non-catalytic ADAMs colon cancer cells, Apoptosis: Int. J. Progr. Cell Death 18 (2013) 1163–1174. are novel integrin α4 ligands, Mol. Cell. Biochem. 442 (2018) 29–38. [66] S. Liu, W. Zhang, K. Liu, B. Ji, G. Wang, Silencing ADAM10 inhibits the in vitro [90] R. Kelwick, I. Desanlis, G.N. Wheeler, D.R. Edwards, The ADAMTS (A disintegrin and in vivo growth of hepatocellular carcinoma cancer cells, Mol. Med. Rep. 11 and metalloproteinase with thrombospondin motifs) family, Genome Biol. 16 (1) (2015) 597–602. (2015) 113. [67] A.V. Jones, D.W. Lambert, P.M. Speight, S.A. Whawell, ADAM10 is over expressed [91] J. Chen, Y. Luo, Y. Zhou, S. Qiu, R. Cui, M. Yu, J. Qin, M. Zhong, Promotion of in oral squamous cell carcinoma and contributes to invasive behaviour through a tumor growth by ADAMTS4 in colorectal cancer: focused on macrophages, Cell. functional association with alphavbeta6 integrin, FEBS (Fed. Eur. Biochem. Soc.) Physiol. Biochem. 46 (4) (2018) 1693–1703. Lett. 587 (2013) 3529–3534. [92] M.O. Kilic, B. Ayenikin, M. Bozer, A. Kara, H. Haltas, D. Icen, K. Demicran, [68] H.B. Liu, Y. Zhu, Q.C. Yang, Y. Shen, X.J. Zhang, H. Chen, Expression and clinical Differentially regulated ADAMTS1, 8, 9, and 18 in pancreas adenocarcinoma, significance of ADAM17 protein in esophageal squamous cell carcinoma, GMR 14 Przeglad Gastroenterol. 12 (4) (2017) 263–270. (2015) 4391–4398. [93] J.S. Fridman, E. Caulder, M. Hansbury, X. Liu, G. Yang, Q. Wang, Y. Lo, B.B. Zhou, [69] X. Zheng, F. Jiang, M. Katakowski, Y. Lu, M. Chopp, ADAM17 promotes glioma A.M. Pan, S.M. Thomas, J.R. Grandis, J. Zhuo, W. Yao, R.C. Newton, cell malignant phenotype, Mol. Carcinog. 51 (2) (2012) 150–164. S.M. Freidman, P.A. Scherle, K. Vaddi, Selective inhibition of ADAM metallopro- [70] G. Sinnathamby, J. Zerfass, J. Hafner, P. Block, Z. Nickens, A. Hobeika, teasesas a novel approach for modulating ErbB pathways in cancer, Clin. Cancer A.A. Secord, H.K. Lyerly, M.A. Morse, R. Philip, ADAM metalloprotease domain 17 Res. 13 (2007) 1892–1902. (ADAM17) is naturally processed through major histocompatibility complex [94] L. Witters, P. Scherle, S. Friedman, J. Fridman, E. Caulder, R. Newton, A. Lipton, (MHC) class 1 molecules and is a potential immunotherapeutic target in breast, Synergistic inhibiton with a dual epidermal growth factor receptor/HER-2/neu ovarian, and prostate cancers, Clin. Exp. Immunol. 163 (3) (2011) 324–332. tyrosine kinase inhibitor and a disintegrin and metalloprotease inhibitor, Can. Res. [71] P.C. Buchanan, K.L.M. Boylan, B. Walcheck, R. Heinze, M.A. Geller, P.A. Argenta, 68 (17) (2008) 7083–7089. A.P.N. Skubitz, Ectodomain shedding of the Nectin-4 in [95] R.C. Newton, E.C. Bradley, R.S. Levy, D. Doval, S. Bondarde, T.P. Sahoo, ovarian cancer is mediated by ADAM10 and ADAM17, J. Biol. Chem. 292 (15) D. Lokanatha, P.K. Julka, R. Nagarkar, S.M. Fridman, Targeting ADAM-mediated (2017) 6339–6351. ligand cleavage to inhibit HER2 and EGFR pathways in non-small cell lung cancer. [72] K.L. Boylan, P.C. Buchanan, R.D. Manion, D.M. Shukla, K. Braumberger, Cancer Cell, 10(1): 39-50 et al. Clinical benefit of INCB7839, a potent and selective C. Bruggemeyer, A.P. Skubitz, The expression of Nectin-4 on the surface of ovarian ADAM inhibitor, in combination with trastuzumab in patients with metastatic cancer cells alters their ability to adhere, migrate, aggregate, and proliferate, HER2C breast cancer, J. Clin. Oncol. 28 (15_suppl) (2006) 3025 supplement, Oncotarget 8 (6) (2017) 9717–9738 7. (2010). [73] M. Ueno, T. Shiomi, S. Mochizuki, M. Chijiiwa, M. Shimoda, Y. Kanai, F. Kataoka, [96] Y. Zhang, M. Hegen, J. Xu, J.C. Keith, G. Jin, X. Du, T. Cummons, B.J. Sheppard, L. A. Hirasawa, N. Susumu, D. Aoki, Y. Okada, ADAM9 is over-expressed in human Sun Y. Zhu, V.R. Rao, Q. Wang, W. Xu, R. Cowling, C.L. Nickerson-Nutter, ovarian clear cell carcinomas and suppresses cisplatin-induced cell death, Cancer J. Gibbons, J. Skotnicki, L.L. Lin, J. Levin J. Characterization of (2R, 3S)-2-([[4-(2- Sci. 109 (2) (2018) 471–482. butynyloxy) phenyl] sulfonyl] amino)-N,3-dihydroxybutanamide, a potent and [74] C.M. Kossmann, M. Annereau, A. Thomas- Schoemann, C. Nicco-Overney, selective inhibitor of TNF-alpha converting enzyme, Int. Immunopharmacol. 4 C. Chereau, F. Batteux, J. Alexandre, F. Lemare, ADAM9 expression promotes an (2004) 1845–1857. aggressive lung adenocarcinoma phenotype, Tumour Biol. 39 (7) (2017) 1–11. [97] P.M. McGowan, C. Simedrea, E.J. Ribot, P.J. Foster, D. Palmieri, P.S. Steeg, [75] V.O. Oria, P. Lopatta, O. Schillig, The pleiotropic roles of ADAM9 in the biology of A.L. Allan, A.F. Chambers, Notch1 inhibition alters the CD44hi/CD24lo popula- solid tumors, Cell. Mol. Life Sci. 75 (13) (2018) 2291–2301. tion and reduces the formation of brain metastases from breast cancer, Mol. [76] V.O. Oria, P. Lopatta, T. Schmitz, B.T. Preca, A. Nyström, C. Conrad, J.W. Bartsch, Cancer Res. 9 (2011) 834–844. B. Kulemann, J. Hoeppner, J. Maurer, P. Bronsert, O. Schilling, ADAM 9 con- [98] X. Lv, Y. Li, M. Qian, C. Ma, H. Jing, Z. Wen, D. Qian, ADAM17 silencing sup- tributes to vascular invasion in pancreatic ductal adenocarcinoma, Mol. Oncol. 13 presses the migration and invasion of non-small cell lung cancer, Mol. Med. Rep. 9 (2019) 456–479. (5) (2014) 1935–1940. [77] K. Iba, R. Albrechtsen, B.J. Gilpin, F. Loechel, U.M. Wewer, Cysteine rich domain [99] C. Hundhausen, D. Misztela, T.A. Berkhout, N. Broadway, P. Saftig, K. Reiss, of human ADAM12 (meltrin alpha) supports tumor cell adhesion, Am. J. Pathol. D. Hartmann, F. Fahrenholz, R. Postina, V. Matthews V, et al., The disintegrin-like 154 (5) (1999) 1489–1501 1999. metalloproteinase ADAM10 is involved in constitutive cleavage of CX3CL1 (frac- [78] S. Shao, Z. Li, W. Gao, G. Yu, D. Liu, F. Pan, ADAM-12 as a diagnostic marker for talkine) and regulates CX3CL1-mediated cell-cell adhesion, Blood 102 (2003) the proliferation, migration and invasion in patients with small cell lung cancer 1186–1195 (A). PLoS One, 9 (Issue 1) (2014) e85936. [100] A. Ludwig, C. Hundhausen, M.H. Lambert, N. Broadway, R.C. Andrews, [79] J. Wang, B. Voellger, J. Benzel, U. Schlomann, C. Nimsky, J.W. Bartsch, B. Carl, D.M. Bickett, M.A. Leesnitzer, J.D. Becherer, Metalloproteinase inhibitors for the Metalloproteinases ADAM12 and MMP-14 are associated with cavernous sinus disintegrin-like ester inducible shedding of cell surface molecules, Comb. Chem. invasion in pituitary adenomas, Int. J. Cancer 139 (6) (2016) 1327–1339. High Throughput Screen. 8 (2005) 161–171. [80] D.J. Cheon, A.J. Li, J.A. Beach, A.E. Walts, H. Tran, J. Lester, B.Y. Karlan, [101] M.L. Moss, A. Stoeck, W. Yan, P.J. Dempsey, ADAM10 as a target for anti-cancer S. Orsulic, ADAM12 is a prognostic factor associated with an aggressive molecular therapy, Curr. Pharmaceut. Biotechnol. 9 (2008) 2–8. subtype of high-grade serous ovarian carcinoma, Carcinogenesis 36 (7) (2015) [102] G. Lorenzatti Hiles, A. Bucheit, J.R. Rubin, A. Hayward, A.L. Cates, K. C Day, L. EI- 739–747. Sawy, L.P. Kunju, S. Daignault, C.T. Lee, M. Leibert, M. Hussain, M.L. Day, [81] N. Lucas, M.L. Day, The role of the disintegrin metalloproteinase ADAM15 in ADAM15 is functionally associated with the metastatic progression of human prostate cancer progression, J. Cell. Biochem. 15 (6) (2009) 967–974 106. bladder cancer, PLoS One 11 (3) (2016) e0150138. [82] V. Beck, H. Herold, A. Benge, B. Luber, P. Hutzler, H. Tschesche, ADAM15 de- [103] F.M. Richards, C.J. Tape, D.I. Jodrell, G. Murphy, Anti-tumour effects of a specific creases integrin alphavbeta3/vitronectin-mediated ovarian cancer cell adhesion anti-ADAM17 antibody in an ovarian cancer model in vivo, PLoS One 7 (7) (2012) and motility in an RGD-dependent fashion, Int. J. Biochem. Cell Biol. 37 (3) (2005) e40597. 590–603. [104] C.J. Tape, S.H. Willems, S.L. Dombernowsky, P.L. Stanley, M. Fogarasi, [83] M. Romagnoli, N.D. Mineva, M. Polmear, C. Conrad, S. Srinivasan, D. Loussouarn, W. Ouwehand, J. McCafferty, G. Murphy, Cross-domain inhibition of TACE ecto- S. Barillé-Nion, I. Georgakoudi, A. Dagg, E.W. McDermott, M.J. Duffy, domain, Proc. Natl. Acad. Sci. U. S. A. 108 (14) (2011) 5578–5583. P.M. McGowan, U. Schlomann, M. Parsons, J.W. Bartsch, G.E. Sonenshein, ADAM8 [105] Y. Huang, N. Benaich, C. Tape, H.F. Kwok, G. Murphy, Targeting the expression in invasive breast cancer promotes tumor dissemination and metas- activity of ADAM17 by an anti-ADAM17 antibody D1 (A12) inhibits head and neck tasis, EMBO Mol. Med. 6 (2) (2014) 278–294. squamous cell carcinoma cell proliferation and motility via blockage of bradykinin [84] U. Schlomann, G. Koller, C. Conrad, T. Ferdous, P. Golfi, A.M. Garcia, S. Barillé- induced HERs transactivation, Int. J. Biol. Sci. 10 (2014) 702–714. Nion, I. Georgakoudi, A. Dagg, E.W. McDermott, M.J. Duffy, P.M. McGowan, [106] J. Rios-Doria, D. Sabol, J. Chesebrough, D. Stewart, L. Xu, R. Tammali, L. Cheng,

56 N. Saha, et al. Cancer Letters 467 (2019) 50–57

Q. Du, K. Schifferli, R. Rothstein, C.C. Leow, J. Heidbrink-Thompson, X. Jin X, Venook X. Wu, J. Ridgway, D. Schahin-Reed, G.J. Dow, A. Shelton, S. Stawicki, C. Gao, J. Friedman, B. Wilkinson, M. Damschroder, A.J. Pierce, R.J. Watts, J. Zhang, R. Choy, P. Howard P, L. Kadyk, M. Yan, J. Zha, C.A. Callahan R.E. Hollingsworth, D.A. Tice, E.F. Michelotti, A monoclonal antibody to ADAM17 CA, S.G. Hymowitz, C.W. Siebel, Therapeutic antibody targeting of individual inhibits tumor growth by inhibiting EGFR and non-EGFR-mediated pathways, Mol. Notch receptors, Nature 464 (2010) 1052–1057 2010. Cancer Ther. 14 (2015) 1637–1649. [114] L. Atapattu, N. Saha, C. Chheang, M.F. Eissman, K. Xu, M.E. Vail, L. Hii, C. Llerena, [107] L. Peng, K. Cook, L. Xu, L. Cheng, M. Damschroder, C. Gao, H. Wu, Z. Liu, K. Horvay, H.E. Abud, U. Kusebauch, R.L. Moritz, B.S. Ding, Z. Cao, S. Rafii, W.F. Dall'Acqua, Molecular basis for the mechanism of action of an anti-TACE M. Ernst, A.M. Scott, D.B. Nikolov, M. Lackman, P.W. Janes, An activated form of antibody, mAbs 8 (8) (2016) 1598–1605. ADAM10 is tumor selective and regulates cancer stem-like cells and tumor growth, [108] K. Yamamoto, A. Trad, A. Baumgart, L. Huske, I. Lorenzen, A. Chalaris, J. Exp. Med. 213 (9) (2016) 1741–1757. J. Grotzinger, T. Dechow, J. Scheller, S. Rose-John, A novel bispecific single-chain [115] T.C. M Seegar, L.B. Killingsworth, N. Saha, P.A. Meyer, D. Patra, B. Zimmerman, antibody for ADAM17 and CD3 induces T-cell-mediated lysis of prostate cancer P.W. Janes, E. Rubinstein, D.B. Nikolov, G. Skiniotis, A.C. Kruse, S.C. Blacklow, cells, Biochem. J. 445 (2012) 135–144. Structural basis for regulated proteolysis by the α-secretase ADAM10, Cell 171 (7) [109] A. Trad A, H.P. Hansen, M. Shomali, M. Peipp, K. Klausz, N. Hedemann, (2017) 1638–1648. K. Yamamoto, A. Mauermann, C. Desel, I. Lorenzen, H. Lemke, S. Rose-John, [116] E. Cerami, J. Gao, U. Dogrusoz, B.E. Gross, S.O. Sumer, B.A. Aksoy, A. Jacobsen, J. Grötzinger, ADAM17-overexpressing breast cancer cells selectively targeted by C.J. Byrne, M.L. Heuer, E. Larsson, Y. Antipin, B. Reva, A.P. Goldberg, C. Sander, antibody-toxin conjugates, Cancer Immunol. Immunother. 62 (3) (2013) 411–421. N. Schultz, The cBio Cancer Genomics Portal: an Open Platform for Exploring [110] J.M. Kim, H.C. Jeung, S.Y. Rha, E.J. Yu, T.S. Kim, Y.K. Shin, K. Yamamoto, Multidimensional Cancer Genomics Data Cancer Discov vol 2, (2012), pp. A. Mauermann, C. Desel, I. Lorenzen, H. Lemke, S. Rose-John, J. Grötzinger, The 401–404. effect of disintegrin-metalloproteinase ADAM9 in gastric cancer progression, Mol. [117] J. Gao, B.A. Aksoy, U. Dogrusoz, G. Dresdner, B. Gross, S.O. Sumer, A. Jacobsen, Cancer Ther. 13 (2014) 3074–3085. C.J. Byrne, M.L. Heuer, E. Larsson, Y. Antipin, B. Reva, A.P. Goldberg, C. Sander, [111] C. Conrad, M. Götte, U. Schlomann, M. Roessler, A. Pagenstecher, P. Anderson, N. Schultz, Integrative analysis of complex cancer genomics and clinical profiles J. Preston, J. Pruessmeyer, A. Ludwig A, R. Li, R.D. Kamm, R. Ritz, B. Carl, using the cBioPortal, Sci. Signal. 6 (2013) pl1. C. Nimsky, J.W. Bartsch, ADAM8 expression in breast cancer derived brain me- [118] P.F. Cheng, R. Dummer, M.P. Levesque, Data mining the Cancer Genome Atlas in tastases: functional implications on MMP-9 expression and transendothelial mi- the era of precision cancer medicine, Swiss Med. Wkly. 145 (2015) w14183. gration in breast cancer cells, Int. J. Cancer 142 (4) (2018) 779–791. [119] S.J. Sirintrapun, A. Zehir, A. Syed, J. Gao, N. Schultz, D.T. Cheng, Translational [112] I. Dikic, M.H. Schmidt, Notch: implications of endogenous inhibitors for therapy, bioinformatics and clinical research (biomedical) informatics, Surg. Pathol. Clin. 8 Bioessays 32 (2010) 481–487. (2015) 269–288. [113] Y. Wu, C. Cain-Hom, L. Choy, T.J. Hagenbeek, G.P. De leon, Y. Chen, D. Finkle, R.

57