Elsevier

Journal of Controlled Release

Volume 338, 10 October 2021, Pages 773-783
Journal of Controlled Release

Release of basic fibroblast growth factor from acoustically-responsive scaffolds promotes therapeutic angiogenesis in the hind limb ischemia model

https://doi.org/10.1016/j.jconrel.2021.09.013Get rights and content

Abstract

Pro-angiogenic growth factors have been studied as potential therapeutics for cardiovascular diseases like critical limb ischemia (CLI). However, the translation of these factors has remained a challenge, in part, due to problems associated with safe and effective delivery. Here, we describe a hydrogel-based delivery system for growth factors where release is modulated by focused ultrasound (FUS), specifically a mechanism termed acoustic droplet vaporization. With these fibrin-based, acoustically-responsive scaffolds (ARSs), release of a growth factor is non-invasively and spatiotemporally-controlled in an on-demand manner using non-thermal FUS. In vitro studies demonstrated sustained release of basic fibroblast growth factor (bFGF) from the ARSs using repeated applications of FUS. In in vivo studies, ARSs containing bFGF were implanted in mice following induction of hind limb ischemia, a preclinical model of CLI. During the 4-week study, mice in the ARS + FUS group longitudinally exhibited significantly more perfusion and less visible necrosis compared to other experimental groups. Additionally, significantly greater angiogenesis and less fibrosis were observed for the ARS + FUS group. Overall, these results highlight a promising, FUS-based method of delivering a pro-angiogenic growth factor for stimulating angiogenesis and reperfusion in a cardiovascular disease model. More broadly, these results could be used to personalize the delivery of therapeutics in different regenerative applications by actively controlling the release of a growth factor.

Introduction

Critical limb ischemia (CLI), an advanced stage of peripheral artery disease (PAD), is the third leading cause of atherosclerotic morbidity [1]. In CLI, arterial blockages reduce blood flow in the extremities, thus leading to chronic pain, numbness, limited mobility and tissue death. The gold standard in CLI treatment is surgical or endovascular revascularization. However, even with intervention, the prognosis for CLI patients is grim. Rates of restenosis can exceed 80% [2] and 29% of patients will either die or undergo a major amputation within one year of diagnosis [3]. Furthermore, 25% of CLI patients are ineligible for revascularization therapy because of existing co-morbidities [4]. Due to these limited options, experimental treatments have sought to improve tissue perfusion by stimulating the growth of new blood vessels to circumvent occluded vessels. One approach that has been evaluated in both preclinical [[5], [6], [7]] and clinical trials [8,9] of PAD/CLI is the use of exogenous, pro-angiogenic growth factors (GFs) such as basic fibroblast growth factor (bFGF). However, conventional administration routes (e.g., intramuscular or intravascular injections) are ineffective for GFs, which have contributed to translational challenges [10].

Hydrogels are widely used within regenerative medicine to deliver GFs [11,12]. These water-laden matrices have been shown to increase the in vivo half-lives of GFs and facilitate angiogenesis. Release of a GF from a hydrogel is dominated by mechanisms like diffusion and matrix degradation [13]. As such, the ability to spatially and/or temporally control GF release from a conventional hydrogel is limited. Comparatively, endogenous pro-angiogenic GFs are expressed in spatiotemporally-regulated patterns during regeneration [14]. Upon implantation of a conventional hydrogel, the kinetics of GF release cannot be actively altered, which hinders the ability to personalize GF therapy based on patient response.

We have developed a composite hydrogel that enables control of GF release using therapeutic, focused US (FUS). Globally, FUS is used clinically in both diagnostic and therapeutic applications [15]. FUS can be applied non-invasively, focused with sub-millimeter precision, and delivered in a spatiotemporally-defined manner to deeply located tissues. Our composite hydrogel, termed an acoustically-responsive scaffold (ARS), consists of a fibrin matrix containing a GF-loaded, phase-shift double emulsion (Fig. 1A) [16,17]. Fibrin, a component of the provisional matrix during wound healing, can facilitate angiogenesis and is approved by the United States Food and Drug Administration as a hemostatic agent [18]. Fibrin is enzymatically degraded within the body by plasmin and matrix metalloproteinases [19]. The phase-shift double emulsion has a water-in-perfluorocarbon (PFC)-in-water (W1/PFC/W2) structure. A water-soluble payload, like a GF, is contained within the innermost W1 phase. The PFC phase, which surrounds the W1 droplets, is highly hydrophobic, thereby acting as a diffusion barrier for the encapsulated GF. PFCs, like perfluoro-n-alkanes, are used in biomedical applications since they are inert, non-metabolizable, and excreted via exhalation [20]. When exposed to FUS, the PFC phase is vaporized due to the tensile (i.e., rarefactional) part of the acoustic wave. This mechanism is known as acoustic droplet vaporization (ADV) and is a threshold-dependent process [21,22]; thus ADV occurs when the applied acoustic pressure (P) is greater than the threshold pressure for ADV (PADV). The double emulsion morphology of the phase-shift emulsion is disrupted by ADV, which releases the encapsulated payload [23]. Thus, ADV enables on-demand release of a GF from an ARS.

Previously, FUS-modulated release of bFGF from an ARS stimulated endothelial network formation in an in vitro co-culture model [24]. Additionally, when ARSs were implanted subcutaneously, FUS-stimulated release of bFGF caused increases in angiogenesis and perfusion [25]. Spatial patterning of ADV in an ARS, and hence bFGF release, was also shown to elicit spatially-directed, host cell migration following in vivo implantation [26].

In prior studies, ARSs were implanted in non-ischemic tissues. Here, we investigate for the first time the impact of bFGF release from an ARS in a murine model of hind limb ischemia (HLI) [27] - a widely-used, preclinical model of CLI (Fig. 1B). bFGF was encapsulated in a phase-shift double emulsion and subsequently incorporated within an ARS. The effect of single versus repeated FUS exposure on bFGF release was initially evaluated in vitro. Subsequently, ARSs were implanted in mice following induction of HLI and ischemia as well as perfusion were longitudinally monitored during the 28-day study. Angiogenesis, fibrosis, and macrophage infiltration were assessed using histological and immunohistochemical staining.

Section snippets

Preparation of phase-shift double emulsion

Phase-shift double emulsions containing perfluorooctane (C8F18, CAS#: 307–34-6, Alfa Aesar, Ward Hill, MA, USA) were prepared using a microfluidic chip (Cat#: 3200146, junction: 14 × 17 μm, Dolomite, Royston, United Kingdom) as previously described [26]. The W1 phase contained 3.6 mg/mL human recombinant bFGF (Cat#: GF003AF, EMD Millipore, Temecula, CA, USA). The double emulsion was characterized in the range of 1–30 μm using a Coulter Counter (Multisizer 4, Beckman Coulter, Brea, CA USA) with

In vitro release of bFGF from ARSs

bFGF release from the ARSs was measured for 14 days (Fig. 2A). Overall, release correlated with the number of times the ARSs were exposed to FUS. Significantly greater release was observed on day 1 for the two groups exposed to FUS versus the ARS control. For ARSs exposed to FUS every three days, significantly greater release was seen beginning on day 5, when compared to the group exposed to FUS only on day 1, at all matched time points. Cmax was 5.8% [5.6, 6.1], 7.6% [7.4, 7.7], and 11.6%

Discussion

Multiple, suprathreshold FUS exposures triggered release of bFGF from ARSs implanted in the HLI model, which led to significantly greater perfusion in the calf muscle, angiogenesis, and recruitment of macrophages as well as less visible ischemia and fibrosis. Overall, these results are consistent with previous studies using the HLI model that showed an inverse correlation between therapeutic efficacy (e.g., angiogenesis, perfusion) and muscle fibrosis [36,37] as well as infiltration of

Conclusions

We have demonstrated that FUS, specifically a mechanism termed ADV, can be used to non-invasively modulate release of bFGF from a fibrin-based ARS. bFGF was initially contained within a phase-shift double emulsion in the ARS. Sustained release of bFGF from the ARS was obtained by applying FUS every three days. FUS-induced release of bFGF yielded significantly greater perfusion in the calf muscle, angiogenesis, and macrophage infiltration compared to other experimental groups. Additionally, less

Acknowledgments

This work was supported by NIH Grant R01HL139656 (M.L.F.). H.J. was supported by funds from Guangzhou First People's Hospital of South China University of Technology. Special thanks to Dr. Allen Brooks (Department of Radiology) for assisting with the synthesis of the fluorosurfactant, Aniket Jivani (Department of Radiology) for designing in CAD the holder for the FUS transducer and the FUS exposure platform, and Dr. William Weadock (Department of Radiology) for helping with 3D printing of CAD

References (63)

  • A. Moncion et al.

    Controlled release of basic fibroblast growth factor for angiogenesis using acoustically-responsive scaffolds

    Biomaterials

    (2017)
  • X. Lu et al.

    Spatially-directed cell migration in acoustically-responsive scaffolds through the controlled delivery of basic fibroblast growth factor

    Acta Biomater.

    (2020)
  • M. Aliabouzar et al.

    Stable and transient bubble formation in acoustically-responsive scaffolds by acoustic droplet vaporization: theory and application in sequential release

    Ultrasonics Sonochemistry

    (2021)
  • X. Lu et al.

    Parametric study of acoustic droplet vaporization thresholds and payload release from acoustically-responsive scaffolds

    Ultrasound Med. Biol.

    (2019)
  • K.A. Garvin et al.

    Controlling the spatial organization of cells and extracellular matrix proteins in engineered tissues using ultrasound standing wave fields

    Ultrasound Med. Biol.

    (2010)
  • N.S. Patil et al.

    Macroporous poly(sucrose acrylate) hydrogel for controlled release of macromolecules

    Biomaterials

    (1996)
  • H.N. Yang et al.

    Transfection of VEGF(165) genes into endothelial progenitor cells and in vivo imaging using quantum dots in an ischemia hind limb model

    Biomaterials

    (2012)
  • R.M. McEnaney et al.

    P2Y2 nucleotide receptor mediates arteriogenesis in a murine model of hind limb ischemia

    J. Vasc. Surg.

    (2016)
  • L. Huang et al.

    Spatially-directed angiogenesis using ultrasound-controlled release of basic fibroblast growth factor from acoustically-responsive scaffolds

    Acta Biomater

    (2021)
  • K.L. Spiller et al.

    The role of macrophage phenotype in vascularization of tissue engineering scaffolds

    Biomaterials

    (2014)
  • S. Barzelai et al.

    Low-intensity ultrasound induces angiogenesis in rat hind-limb ischemia

    Ultrasound Med. Biol.

    (2006)
  • A.A. Hellingman et al.

    Variations in surgical procedures for hind limb Ischaemia mouse models result in differences in collateral formation

    Eur J Vasc Endovasc

    (2010)
  • A. Moncion et al.

    Sequential payload release from acoustically-responsive scaffolds uinsg focused ultrasound

    Ultrasound Med. Biol.

    (2018)
  • J.A. Mustapha et al.

    Determinants of long-term outcomes and costs in the Management of Critical Limb Ischemia: a population-based cohort study

    J. Am. Heart Assoc.

    (2018)
  • M. Meloni et al.

    Characteristics and outcome for persons with diabetic foot ulcer and no-option critical limb ischemia

    J Clin Med

    (2020)
  • O. Jeon et al.

    Control of basic fibroblast growth factor release from fibrin gel with heparin and concentrations of fibrinogen and thrombin

    J. Control. Release

    (2005)
  • H.S. Yang et al.

    Delivery of basic fibroblast growth factor using heparin-conjugated fibrin for therapeutic angiogenesis

    Tissue Eng Pt A

    (2010)
  • K. Ono et al.

    First-in-Man Clinical Pilot Study Showing the Safety and Efficacy of Intramuscular Injection of Basic Fibroblast Growth Factor With Atelocollagen Solution for Critical Limb Ischemia

    Circulation Journal

    (2019)
  • J.P. Cooke et al.

    Vascular regeneration in peripheral artery disease

    Arterioscler. Thromb. Vasc. Biol.

    (2020)
  • J.Y. Li et al.

    Designing hydrogels for controlled drug delivery

    Nat Rev Mater

    (2016)
  • S. Mantha et al.

    Smart hydrogels in tissue engineering and regenerative medicine

    Materials (Basel)

    (2019)
  • Cited by (23)

    View all citing articles on Scopus
    View full text