Elsevier

Biomaterials

Volume 103, October 2016, Pages 101-115
Biomaterials

Cathepsin S-cleavable, multi-block HPMA copolymers for improved SPECT/CT imaging of pancreatic cancer

https://doi.org/10.1016/j.biomaterials.2016.05.036Get rights and content

Abstract

This work continues our efforts to improve the diagnostic and radiotherapeutic effectiveness of nanomedicine platforms by developing approaches to reduce the non-target accumulation of these agents. Herein, we developed multi-block HPMA copolymers with backbones that are susceptible to cleavage by cathepsin S, a protease that is abundantly expressed in tissues of the mononuclear phagocyte system (MPS). Specifically, a bis-thiol terminated HPMA telechelic copolymer containing 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) was synthesized by reversible addition−fragmentation chain transfer (RAFT) polymerization. Three maleimide modified linkers with different sequences, including cathepsin S degradable oligopeptide, scramble oligopeptide and oligo ethylene glycol, were subsequently synthesized and used for the extension of the HPMA copolymers by thiol–maleimide click chemistry. All multi-block HPMA copolymers could be labeled by 177Lu with high labeling efficiency and exhibited high serum stability. In vitro cleavage studies demonstrated highly selective and efficient cathepsin S mediated cleavage of the cathepsin S-susceptible multi-block HPMA copolymer. A modified multi-block HPMA copolymer series capable of Förster Resonance Energy Transfer (FRET) was utilized to investigate the rate of cleavage of the multi-block HPMA copolymers in monocyte-derived macrophages. Confocal imaging and flow cytometry studies revealed substantially higher rates of cleavage for the multi-block HPMA copolymers containing the cathepsin S-susceptible linker. The efficacy of the cathepsin S-cleavable multi-block HPMA copolymer was further examined using an in vivo model of pancreatic ductal adenocarcinoma. Based on the biodistribution and SPECT/CT studies, the copolymer extended with the cathepsin S susceptible linker exhibited significantly faster clearance and lower non-target retention without compromising tumor targeting. Overall, these results indicate that exploitation of the cathepsin S activity in MPS tissues can be utilized to substantially lower non-target accumulation, suggesting this is a promising approach for the development of diagnostic and radiotherapeutic nanomedicine platforms.

Introduction

In 2015, pancreatic cancer was estimated to be the 11th and 8th most commonly diagnosed cancer for men and women in the United States, respectively. At the same time, it is estimated to account as the 4th leading cause of cancer related death for both men and women [1]. Pancreatic ductal adenocarcinoma (PDAC), which constitutes over 90% of pancreatic cancers in humans, is a devastating and virtually unexceptionally lethal malignancy [2]. Due to a lack of symptoms in early stages of the disease, PDAC is typically not diagnosed until the disease has progressed to a more advanced state [3], [4]. As a result, despite advances in the field of surgery, chemotherapy, and radiation therapy, the prognosis of PDAC remains extremely poor with a five-year survival rate of less than 5% [5], [6], which is the lowest among common malignancies. Because of the high mortality and rapid progression associated with PDAC, effective detection and accurate staging of pancreatic cancer have become a major challenge in disease management.

Over the years, numerous imaging modalities have been utilized to non-invasively detect cancer. In particular, magnetic resonance imaging [7], positron emission tomography [8], single-photon emission computed tomography (SPECT) [9], computed tomography (CT) [10] and ultrasound [11] have played critical roles in improving the diagnosis and staging of many malignancies. As one of the major imaging techniques used in nuclear medicine today, SPECT is often combined with CT to allow co-registration of the radiotracer and anatomy [12]. Utilizing the dual-modality of SPECT/CT has been shown to yield improved sensitivity and accuracy in cancer imaging [13]. Unfortunately, the potential advantages of SPECT/CT imaging for the detection and staging of PDAC cannot currently be realized due to the lack of a clinically approved imaging agent. As a result, the preclinical development of SPECT imaging agents for pancreatic cancer has been an active area of research [14], [15], [16]. These radiotracers have utilized carriers that range from small molecules to macromolecules. To date, the bulk of the macromolecular work has focused on antibodies due to their excellent in vivo targeting and tumor retention properties. Despite the comparable tumor uptake and retention properties, nanomedcines have received relatively little attention in the development of SPECT imaging agents for PDAC and other cancers. This is largely attributable to the high blood retention and uptake of many of these platforms in non-target tissues associated with the mononuclear phagocyte system (MPS) [17]. In order for nanomedicines to become more attractive as a radionuclide carrier, approaches are needed to lower this non-target retention.

During the last two decades, work by Kopeček and other laboratories have demonstrated the potential of poly[N-(2-hydroxypropyl)methacrylamide] (HPMA), a water-soluble, non-immunogenic, non-toxic polymeric platform, as a drug carrier [18]. For cancer, HPMA copolymers have shown great potential as a drug delivery system to enhance the delivery of chemotherapeutics to a variety of tumors through the enhanced permeability and retention (EPR) effect [19]. Although these copolymers exhibited remarkable advantages for drug delivery, the accumulation of this polymeric platform, as with many nanomedicines, in MPS-associated tissues (e.g., blood, liver and spleen) diminishes its potential as a diagnostic agent. Our laboratory has been focused on developing approaches to substantially improve the clearance of radiolabeled HPMA copolymers from MPS-associated, non-target tissues. Our efforts have focused on cathepsin S, a protease that is abundantly expressed in antigen presenting cells (APCs) such as monocytes and macrophages - the primary components of the MPS [20], [21]. Utilizing known peptidic substrates of cathepsin S, we have used these peptides to substantially enhance the clearance of radiolabeled HPMA copolymers from MPS-associated tissues, such as the blood, liver and spleen [22], [23].

Inspired by the recent work on backbone biodegradable copolymers [24], [25], this manuscript investigates the incorporation of cathepsin S susceptible linkers into the HPMA backbone and its impact on the biological performance of these agents for PDAC imaging. We hypothesized that inclusion of the cathepsin S cleavable, peptidic linker would result in fragmentation of the HPMA copolymers and faster clearance of the copolymer fragments from phagocytic cells (e.g., monocytes and macrophages) and tissues associated with MPS uptake and retention. Furthermore, as a consequence of this fragmentation, the smaller copolymer blocks would be expected to clear more quickly from the body. To investigate, we designed two multi-block HMPA copolymer platforms, depicted in Fig. 1: MPs and FMPs. The MPs, which contains the chelation moiety, were radiolabeled with lutetium-177 (177Lu) and evaluated using various in vitro and in vivo studies. Whereas, the FMPs, which are capable of Förster Resonance Energy Transfer (FRET), were utilized as a tool to investigate in vitro fragmentation kinetics of the biodegradable, multi-block HPMA copolymers. For both the MPs and FMPs, a known cathepsins S peptide substrate (PMGLP) was utilized as the linker as well as controls containing a scrambled peptide analogue and a non-cleavable polyethylene glycol (PEG). Herein, the syntheses, characterization, in vitro cleavage assays and cell trafficking studies for the MPs and FMPs are described. In addition, the biodistribution studies and SPECT/CT imaging results of the MPs in a PDAC mouse model are reported.

Section snippets

Materials

Reagent: All solvents used for reaction and silica gel purification were ACS grade and purchased from Fisher Scientific. Acetonitrile was HPLC grade and purchased from Fisher Scientific. Water was deionized by Millipore® Milli Q Biocell Ultrapure Water System before use. CDCl3 and deuterium oxide were obtained from ACROS Organics™. Fluorescein isothiocyanate (FITC), N-Boc-ethylenediamine, N-hydroxysuccinimide (NHS), N,N′-dicyclohexylcarbodiimide (DCC), natLutetium trichloride,

Synthesis of CTAs and initiator

As depicted in Fig. 1, two types of multi-block HPMA copolymers, MPs and FMPs, were targeted for synthesis, synthetic schemes given in Scheme 1, Scheme 2, respectively. The syntheses of the telechelic-block copolymers BP and FBP were envisioned to be carried out by RAFT polymerization [30], [31], [32]. In order to carry out this synthesis, chain transfer agents (CTA) and initiators with terminal thiol groups needed to be synthesized for each scheme. The structures of the designed CTAs and

Conclusion

In this study, two groups of backbone cathepsin S-susceptible HPMA copolymers (MPs and FMPs) were synthesized and characterized for in vitro and in vivo evaluation. Utilization of the cathepsin S-susceptible linkers gave multi-block copolymers with rapid in vitro cleavage kinetics for both the MP-c and FMP-c copolymers. This was further confirmed using in vivo biodistribution and SPECT/CT imaging studies in a PDAC mouse model. Relative to controls, 177Lu-MP-c demonstrated substantially higher

Acknowledgments

We thank Janice A. Taylor and James R. Talaska from the Advanced Microscopy Core Facility at the UNMC for providing assistance with (confocal or super resolution) microscopy; Philip Hexley, Victoria B. Smith and Samantha Wall from the flow cytometry research facility at the UNMC for providing technical support in flow cytometry studies; Li Wu and Na Ly from the Elutriation Core Facility of UNMC for the technical support in cell study; Yuning Zhang from College of Medicine at the UNMC for

References (54)

  • R.N. Johnson et al.

    HPMA-oligolysine copolymers for gene delivery: optimization of peptide length and polymer molecular weight

    J. Control. Release

    (2011)
  • V. Omelyanenko et al.

    Targetable HPMA copolymer-adriamycin conjugates. Recognition, internalization, and subcellular fate

    J. Control. Release

    (1998)
  • J. Kopecek

    Polymer-drug conjugates: origins, progress to date and future directions

    Adv. Drug Deliv. Rev.

    (2013)
  • J. Shi et al.

    Engineering biodegradable and multifunctional peptide-based polymers for gene delivery

    J. Biol. Eng.

    (2013)
  • G.-P. Shi et al.

    Human cathepsin S: chromosomal localization, gene structure, and tissue distribution

    J. Biol. Chem.

    (1994)
  • H.R. Kruger et al.

    Imaging of doxorubicin release from theranostic macromolecular prodrugs via fluorescence resonance energy transfer. Journal of controlled release

    Off. J. Control. Release Soc.

    (2014)
  • S. Petanceska et al.

    Expression of rat cathepsin S in phagocytic cells

    J. Biol. Chem.

    (1996)
  • S.W. Waldo et al.

    Heterogeneity of human macrophages in culture and in atherosclerotic plaques

    Am. J. Pathol.

    (2008)
  • H. Maeda et al.

    Tumor vascular permeability and the EPR effect in macromolecular therapeutics: a review

    J. Control. Release

    (2000)
  • R.L. Siegel et al.

    Cancer statistics

    CA Cancer J. Clin.

    (2015)
  • A.V. Biankin et al.

    Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes

    Nature

    (2012)
  • K. Hanada et al.

    Diagnostic strategies for early pancreatic cancer

    J. Gastroenterol.

    (2015)
  • I.H. Sahin et al.

    Molecular signature of pancreatic adenocarcinoma: an insight from genotype to phenotype and challenges for targeted therapy

    Expert Opin. Ther. Targets

    (2015)
  • R. Siegel et al.

    Cancer statistics, 2011: the impact of eliminating socioeconomic and racial disparities on premature cancer deaths

    CA Cancer J. Clin.

    (2011)
  • J. Grimm et al.

    Magnetic resonance imaging of the pancreas and pancreatic tumors in a mouse orthotopic model of human cancer

    Int. J. Cancer J. Int. Cancer

    (2003)
  • M.T. Madsen

    Recent advances in SPECT imaging

    J. Nucl. Med

    (2007)
  • K.S. Gurusamy et al.

    Diagnostic accuracy of different imaging modalities following computed tomography (CT) scanning for assessing the resectability with curative intent in pancreatic and periampullary cancer

    Cochrane Libr.

    (2015)
  • Cited by (24)

    • Protease-triggered bioresponsive drug delivery for the targeted theranostics of malignancy

      2021, Acta Pharmaceutica Sinica B
      Citation Excerpt :

      In view of this, except as a therapeutic target, the properties of cathepsin S have been integrated into nanocomplexes for protease-susceptible oncologic imaging or targeted therapy. In a typical manner, Fan et al.230 synthesized a cathepsin S-degradable multi-block copolymers based on a water-soluble and biofriendly polymer, poly[N-(2-hydroxypropyl)methacrylamide] (HPMA), as the backbone. By coupling cathepsin S-sensitive peptide substrate (PMGLP) and radiolabeling with lutetium-177 (177Lu), the nanocomposites showed highly sensitive single-photon emission computed tomography (SPECT)/CT imaging in PDAC.

    • Fabrication of versatile dynamic hyaluronic acid-based hydrogels

      2020, Carbohydrate Polymers
      Citation Excerpt :

      The encapsulation of nanoparticles, microspheres, or liposomes in our hydrogel might be better options, given their relatively large size, and they might be released in a more sustained manner (Burade et al., 2017; Wang et al., 2011). Second, like the problem associated with the host-guest HA-based hydrogel, the relaxation after extrusion of the hydrogel during 3D printing easily de-stabilized the scaffold structure, suggesting that a secondary crosslinking strategy compatible with our hydrogel system, e.g. thiol-maleimide click chemistry, should be incorporated to fabricate better constructs for future applications (W. Fan et al., 2016; Jansen, Negron-Pineiro, Galarza, & Peyton, 2018; Ouyang et al., 2016). In summary, we successfully and facilely synthesized dynamic HA-based hydrogels with self-healing and ROS-responsive properties that are relatively stable in physiologically relevant conditions.

    • Polymer nanomedicines

      2020, Advanced Drug Delivery Reviews
      Citation Excerpt :

      However, other factors, such as the conformation of the macromolecule [66,96,97], association of side-chains by hydrophobic interactions (formation of unimolecular micelles), or random association of flexible chains by “point-point” contacts [98–100], influence the rate of enzymatic drug release [37,38], penetration of the solid tumor [101] and, ultimately, the efficacy. Similar approach based on RAFT polymerization and incorporation of enzymatically degradable sequences into the polymer backbone was used by Luo’s [102–104] and Garrison’s labs [43–45,105]. Pechar et al. used different chemistry; they prepared multiblock copolymers by interfacial polycondensation of telechelic PEG (2 kDa) and oligopeptide diamine from glutamic acid and lysine.

    • Urokinase injection-triggered clearance enhancement of a 4-arm PEG-conjugated <sup>64</sup>Cu-bombesin analog tetramer: A novel approach for the improvement of PET imaging contrast

      2018, International Journal of Pharmaceutics
      Citation Excerpt :

      Using the methodology presented here, we are currently examining imaging contrast improvement for antibody PET probes (immuno-PET), which is expected to improve small chemical tracer specificity (Tamura et al., 2013; Jauw et al., 2016). Recently, peptide-cleavable linkers by endogenous enzymes such as cathepsin S were used to improve tumor to tissue contrast in N-(2-hydroxypropyl)methacrylamide copolymer-single photon emission computerized tomography (Fan et al., 2016; Fan et al., 2017; Zhang et al., 2013). These linkers were cleaved in mononuclear phagocyte system associated tissues, including the liver and spleen, which reduced nontarget accumulation.

    View all citing articles on Scopus
    1

    These authors contributed equally to this work.

    View full text