Development of an optimized activatable MMP-14 targeted SPECT imaging probe

https://doi.org/10.1016/j.bmc.2008.11.078Get rights and content

Abstract

Matrix metalloproteinase-14 (MT1-MMP or MMP-14) is a membrane-associated protease implicated in a variety of tissue remodeling processes and a molecular hallmark of select metastatic cancers. The ability to detect MMP-14 in vivo would be useful in studying its role in pathologic processes and may potentially serve as a guide for the development of targeted molecular therapies. Four MMP-14 specific probes containing a positively charged cell penetrating peptide (CPP) d-arginine octamer (r8) linked with a MMP-14 peptide substrate and attenuating sequences with glutamate (8e, 4e) or glutamate-glycine (4eg and 4egg) repeating units were modeled using an AMBER force field method. The probe with 4egg attenuating sequence exhibited the highest CPP/attenuator interaction, predicting minimized cellular uptake until cleaved. The in vitro MMP-14-mediated cleavage studies using the human recombinant MMP-14 catalytic domain revealed an enhanced cleavage rate that directly correlated with the linearity of the embedded peptide substrate sequence. Successful cleavage and uptake of a technetium-99m labeled version of the optimal probe was demonstrated in MMP-14 transfected human breast cancer cells. Two-fold reduction of cellular uptake was found in the presence of a broad spectrum MMP inhibitor. The combination of computational chemistry, parallel synthesis and biochemical screening, therefore, shows promise as a set of tools for developing new radiolabeled probes that are sensitive to protease activity.

Graphical abstract

Design strategy combining computational chemistry, parallel synthesis and protease cleavage studies has yielded a promising MMP-14 sensitive probe for SPECT imaging.

  1. Download : Download full-size image

Introduction

Tumor cells employ a host of biochemical mechanisms in order to invade and metastasize. Many of these mechanisms are thought, in part, to involve proteases associated with cell membrane and extracellular matrix (ECM) molecules that are posited to initiate pro-angiogenic signaling cascades. Among cancer-associated proteases, matrix metalloproteases (MMPs), a class of zinc-dependent proteolytic enzymes, have been postulated to be used by cancer cells to dissolve ECM during neoplastic progression.1 In addition, numerous studies have documented a positive correlation between certain MMP expression levels and poor outcome in cancer patients.2 The importance of MMPs in tumor progression not only has guided the development of MMP inhibitors for therapy, it has also received particular attention as imaging target utilizing methods to detect tumor-associated proteolytic activity in vivo.3, 4, 5, 6, 7

The family of human MMPs contains 16 secreted and 7 membrane-tethered enzymes.8 A subclass of the membrane-anchored proteinases, termed membrane type (MT) MMPs, plays dominant roles in controlling cancer cell behavior.9, 10 In particular, the up-regulation of the membrane-associated collagenase MMP-14 (MT1-MMP) correlates to the invasiveness of many different tumor types.2 MMP-14 not only promotes tumor growth through induction of angiogenesis and proteolysis of ECM, but it also acts as a critical regulatory switch in the activation of MMP-2 proenzyme.11 Clinical studies revealed that the expression of MMP-14 is associated with poor prognosis in patients with advanced neuroblastoma,12 small cell lung cancer (SCLC),13 tongue squamous cell carcinoma,14 head and neck carcinoma,15 bladder, and ovarian cancer.16, 17 MMP-14 has been detected in tumor cells and adjacent stromal cells in a variety of human tumors including breast.9 Consequently, MMP-14 overexpression holds great promise as an early biomarker for invasive cancers.

In the past, several in vivo optical imaging probes targeting various MMPs have been reported; the most successful of these efforts have been directed against MMP-2, -7, and -9.18, 19, 20, 21, 22, 23 Attempts to image MMP activities by non-optical modalities (e.g., positron emission tomography (PET) or single photon emission computed tomography (SPECT)) using labeled substrates or inhibitors, however, have met with limited success in vivo, in part due to the poor specificity and in vivo stability of the radiolabeled probes.24, 25, 26, 27, 28, 29, 30, 31 Our motivation, therefore, was to develop a sensitive nuclear probe for MMP-14 activities for early cancer detection. The success of such a probe would represent a significant advancement in preclinical and clinical imaging as it would be a tool able to locate and track the molecular evolution of malignant tissues for use in drug development.

A number of protease imaging strategies have been described previously. One particular class of probes comprising an ‘activatable’ delivery mechanism has been developed by a number of research groups.21, 23, 32 These probes share a core structure consisting of a poly-d-arginine cell penetrating peptide (CPP) that is covalently tethered to a negatively charged attenuating peptide sequence through a proteolysis sensitive peptide (Fig. 1). The intact probe is believed to be prohibited from crossing the cell membrane due to the electrostatic interaction between the positively charged arginine and the tethered intramolecular negatively charged attenuator (I), but other mechanisms may also contribute. However, proteolytic cleavage (II) separates the polyarginine sequence from the negatively charged domain, thereby triggering uptake of the CPP (III). Protease-rich tissues may be imaged by tagging an imaging reporter group, such as a fluorophore, gadolinium chelate or radionuclide, to the CPP.32 As a result, the number of protease cleavage events may be correlated to the CPP concentration, and its associated tag, within targeted cells. Using agents directed against MMP-2/9 and -7, Rao and co-workers have selectively tagged cultured fibrosarcoma cells (HT-1080) with quantum dots,23 while Tsien and co-workers successfully imaged cancers rich in MMP-2 and -9 in murine xenografts, using optical and magnetic resonance techniques.21

This general strategy is attractive because the catalytic processing of more than one probe by each enzyme provides a robust mechanism for signal amplification. For the purpose of MMP-14 imaging, this is a particularly important point if one seeks to detect protease activity prior to the maturation of secondary downstream proteases, for example, MMP-2 and -9.

The development of an ‘activatable’ SPECT imaging probe specific for MMP-14 is reported herein. The probe design was undertaken realizing that attaching a large metal chelate for nuclear imaging may alter the topology of the MMP-14 selective peptide sequence and adversely affect the cleavage rate as well as attenuation characteristics of the basic probe platform. Therefore the combination of molecular modeling, parallel synthesis and bioassay screens were effectively utilized to optimize the imaging probe construct for MMP-14 activities. This work sheds new light on the intramolecular quenching and activation mechanisms of this class of imaging peptides, and demonstrates the value of computational chemistry relative to imaging probe development.

Section snippets

MMP-14 probe design and modular components

The general MMP-14 probe, shown in Figure 2, is a modular design comprised of three components: (A) a positively charged d-arginine octamer (r8) cell penetrating peptide (CPP) attached with single amino acid chelate (SAAC) for technetium-99m; (B) a MMP-14 specific cleavable substrate (SGRIGF↓LRTA) and (C) a negatively charged attenuation sequence. The seminal component for designing an effective MMP-14 probe was a suitable cleavable peptide substrate. A known MMP-14 substrate (SGRIGF↓LRTA) was

Conclusion

The unique application of molecular modeling in optimization of the MMP-14 probe construct has been demonstrated. Modeling calculations were able to provide useful information on the attenuation properties of four different sequences against the r8 CPP and the length of the MMP substrate that is directly related to cleavage rate. Both of attenuation and cleavage must be maximized in the optimal probe. Although three out of four probes studied exhibited good CPP attenuation, the CPP/attenuator

Molecular modeling

Computer simulations were used to predict the solution structures of the peptide probes employing the AMBER96 force field42 with the implicit solvation model of Onufriev, Bashford, and Case.43 This force field was validated in a separate study and shown to give accurate structures for peptides with both alpha and beta motifs.44 For the non-canonical chelate residue, the program ‘antechamber’ in the AMBER package was used to determine atomic charges and estimate force field constants. The SAAC

Acknowledgements

This work was supported by the Office of Science, Office of Biological and Environmental Research, Biological Systems Science Division, U.S. Department of Energy (DE-FG02-05ER64010). G.W. was supported by a Department of Defense Breast Cancer Center of Excellence grant DAMD17-02-1-0693, and L.M.C. acknowledges R01 support from the NIH/NCI and a DOD BCRP Era of Hope Scholar Award (W81XWH-06-1-0416).The authors wish to thank Molecular Insight Pharmaceuticals, Inc. for their gift of the

References and notes (49)

  • M. Seiki

    Cancer Lett.

    (2003)
  • S. Zucker et al.

    Curr. Top. Dev. Biol.

    (2003)
  • N.E. Sounni et al.

    Biochimie

    (2005)
  • A.Y. Strongin et al.

    J. Biol. Chem.

    (1995)
  • B. Davidson et al.

    Mol. Cell Endocrinol.

    (2002)
  • S. Furumoto et al.

    Nucl. Med. Biol.

    (2003)
  • K. Kopka et al.

    Nucl. Med. Biol.

    (2004)
  • R. Oltenfreiter et al.

    Nucl. Med. Biol.

    (2004)
  • J.E. Sprague et al.

    Nucl. Med. Biol.

    (2006)
  • Q.H. Zheng et al.

    Nucl. Med. Biol.

    (2002)
  • S.J. Kridel et al.

    J. Biol. Chem.

    (2002)
  • Y. Sugita et al.

    Chem. Phys. Lett.

    (1999)
  • P.D. Gupta et al.

    FEBS Lett.

    (1992)
  • M.D. Sternlicht et al.

    Oncogene

    (2000)
  • W. Cai et al.

    Mol. Cancer Ther.

    (2006)
  • C.M. Overall et al.

    Nat. Rev. Cancer

    (2002)
  • R.L. Scherer et al.

    Cancer Metastasis Rev.

    (2008)
  • C. Van de Wiele et al.

    Cancer Biother. Radiopharm.

    (2006)
  • S. Zucker et al.

    Nat. Med.

    (2001)
  • A. Page-McCaw et al.

    Nat. Rev. Mol. Cell Biol.

    (2007)
  • M. Sakakibara et al.

    Cancer

    (1999)
  • M. Michael et al.

    J. Clin. Oncol.

    (1999)
  • T. Yoshizaki et al.

    Int. J. Cancer

    (2001)
  • T. Yoshizaki et al.

    Cancer

    (1997)
  • Cited by (57)

    • The functional cross talk between cancer cells and cancer associated fibroblasts from a cancer mechanics perspective

      2021, Biochimica et Biophysica Acta - Molecular Cell Research
      Citation Excerpt :

      The secretion of MMPs by CAFs is a determinant of ECM remodeling leading to invasion [26,229,264,303], by providing necessary pathways through activation of Rho-ROCK cascade and proteolytic activity, which are tracks for migration of CAFs and cancer cells individually or collectively [304]. In a wide range of tissues, the high expression of MMP family has been shown to be associated with cancer progress [305–307], and overexpression of MMPs is a sign of metastatic cascade [308–311]. The secretion of MMPs paves the way for cancer cell invasion through the basement membrane and endothelium which is necessary during metastatic intravasation [312].

    • Challenge to overcome current limitations of cell-penetrating peptides

      2021, Biochimica et Biophysica Acta - Proteins and Proteomics
    • Exploiting proteases for cancer theranostic through molecular imaging and drug delivery

      2020, International Journal of Pharmaceutics
      Citation Excerpt :

      The technetium-99 m labelled probe containing charged cell-penetrating peptide, d-arginine octamer (r8) conjugated with MMP-14 peptide sequence was synthesized. SPECT imaging with the optimal probe with the longest glutamate-glycine sequence showed successful cleavage uptake in MMP-14 transfected human breast cancer cells (Watkins et al., 2009). Coumarin-based FRET probe was also synthesized in which a polypeptide chain link was formed between the fluorophore 7-[diethylamino]-2-oxo-2H-chromene-3- succinimidyl ester (Coumarin-Osu) and the quencher 4-(4-[dimethylamino]phenylazo) benzoic acid N-succinimidyl ester (Dabcyl- Osu).

    • Divergent changes in the content and activity of MMP-26 and TIMP-4 in human umbilical cord tissues associated with preeclampsia

      2018, European Journal of Obstetrics and Gynecology and Reproductive Biology
      Citation Excerpt :

      Assay was performed in black 96 flat bottom well microplate, which was pre-coated with specific MMP antibody (the same antibody that was used in Western blot analysis) for immobilization of the metalloproteinase [17]. MMP activity was measured according to Watkins et al. [18] with MCA-Pro-Leu-Ala-Cys(p-OMeBz)-Trp-Ala-Arg(Dpa)-H2 (Merck, cat. no 444,258) as a fluorogenic substrate.

    View all citing articles on Scopus
    View full text