Safety and feasibility of ultrasound-triggered targeted drug delivery of doxorubicin from thermosensitive liposomes in liver tumours (TARDOX): a single-centre, open-label, phase 1 trial

Paul C Lyon, Michael D Gray, Christophoros Mannaris, Lisa K Folkes, Michael Stratford, Leticia Campo, Daniel Y F Chung, Shaun Scott, Mark Anderson, Robert Goldin, Robert Carlisle, Feng Wu, Mark R Middleton, Fergus V Gleeson, Constantin C Coussios, Paul C Lyon, Michael D Gray, Christophoros Mannaris, Lisa K Folkes, Michael Stratford, Leticia Campo, Daniel Y F Chung, Shaun Scott, Mark Anderson, Robert Goldin, Robert Carlisle, Feng Wu, Mark R Middleton, Fergus V Gleeson, Constantin C Coussios

Abstract

Background: Previous preclinical research has shown that extracorporeal devices can be used to enhance the delivery and distribution of systemically administered anticancer drugs, resulting in increased intratumoural concentrations. We aimed to assess the safety and feasibility of targeted release and enhanced delivery of doxorubicin to solid tumours from thermosensitive liposomes triggered by mild hyperthermia, induced non-invasively by focused ultrasound.

Methods: We did an open-label, single-centre, phase 1 trial in a single UK hospital. Adult patients (aged ≥18 years) with unresectable and non-ablatable primary or secondary liver tumours of any histological subtype were considered for the study. Patients received a single intravenous infusion (50 mg/m2) of lyso-thermosensitive liposomal doxorubicin (LTLD), followed by extracorporeal focused ultrasound exposure of a single target liver tumour. The trial had two parts: in part I, patients had a real-time thermometry device implanted intratumourally, whereas patients in part II proceeded without thermometry and we used a patient-specific model to predict optimal exposure parameters. We assessed tumour biopsies obtained before and after focused ultrasound exposure for doxorubicin concentration and distribution. The primary endpoint was at least a doubling of total intratumoural doxorubicin concentration in at least half of the patients treated, on an intention-to-treat basis. This study is registered with ClinicalTrials.gov, number NCT02181075, and is now closed to recruitment.

Findings: Between March 13, 2015, and March 27, 2017, ten patients were enrolled in the study (six patients in part I and four in part II), and received a dose of LTLD followed by focused ultrasound exposure. The treatment resulted in an average increase of 3·7 times in intratumoural biopsy doxorubicin concentrations, from an estimate of 2·34 μg/g (SD 0·93) immediately after drug infusion to 8·56 μg/g (5·69) after focused ultrasound. Increases of two to ten times were observed in seven (70%) of ten patients, satisfying the primary endpoint. Serious adverse events registered were expected grade 4 transient neutropenia in five patients and prolonged hospital stay due to unexpected grade 1 confusion in one patient. Grade 3-4 adverse events recorded were neutropenia (grade 3 in one patient and grade 4 in five patients), and grade 3 anaemia in one patient. No treatment-related deaths occurred.

Interpretation: The combined treatment of LTLD and non-invasive focused ultrasound hyperthermia in this study seemed to be clinically feasible, safe, and able to enhance intratumoural drug delivery, providing targeted chemo-ablative response in human liver tumours that were refractory to standard chemotherapy.

Funding: Oxford Biomedical Research Centre, National Institute for Health Research.

Copyright © 2018 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY license. Published by Elsevier Ltd.. All rights reserved.

Figures

Figure 1
Figure 1
Trial profile
Figure 2
Figure 2
Total doxorubicin concentration in plasma and tumour samples analysed by high-performance liquid chromatography (HPLC) (A) Lyso-thermosensitive liposomal doxorubicin (LTLD) plasma pharmacokinetic data by HPLC. Data for patient I.01 are omitted from the plot, because concentrations were much greater than the top standard, resulting in a ten-fold dilution step for plasma analysis subsequently being introduced to the assay. (B) Intratumoural pharmacokinetic data by HPLC. The post-LTLD values for patient I.02 and I.06, and the post-LTLD plus focused ultrasound (FUS) values for patient I.06 are worst-case estimates.
Figure 3
Figure 3
Illustrative controlled hyperthermia by focused ultrasound Real-time thermometry data (trace) captured after infusion of lyso-thermosensitive liposomal doxorubicin (LTLD) and during focused ultrasound exposure in moving beam (linear) mode for patient I.05. This trace was acquired at a 10 ms resolution by use of a calibrated Medtronic thermocouple, with custom LabView data-acquisition setup. Shaded regions represent the period when focused ultrasound was being applied. From approximately 30 s to 33 min, a 90·9 cm3 prescribed target tumour volume was exposed to focused ultrasound at 115 W (8·7 MPa peak rarefactional in situ pressure) at 70% duty cycle in linear mode. Although the release threshold was reached within 5 min of focused ultrasound exposure, heating in the first 30 min was deemed slightly suboptimal because of prolonged cooling periods between treatment cycles. Subsequently, by removing the outermost slices from the prescribed treatment volume, resulting in a smaller 68·3 cm3 tumour volume, and increasing power to 125 W (9·0 MPa derated) and duty cycle to 77%, optimal hyperthermia was achieved for 35–80 min. Once focused ultrasound stopped, the tumour was allowed to cool before the thermocouple was removed from the patient at 85 min, and a tumour biopsy sample was subsequently taken. The dotted curve is a fourth order polynomial fit, which is probably more representative of the bulk temperature in the prescribed tumour volume than the rapidly fluctuating point temperature recorded by the sensitive region of the intratumoural thermometry device (trace).

References

    1. Trédan O, Galmarini CM, Patel K, Tannock IF. Drug resistance and the solid tumor microenvironment. J Natl Cancer Inst. 2007;99:1441–1454.
    1. Diederich CJ, Hynynen K. Ultrasound technology for hyperthermia. Ultrasound Med Biol. 1999;25:871–887.
    1. Mura S, Nicolas J, Couvreur P. Stimuli-responsive nanocarriers for drug delivery. Nat Mater. 2013;12:991–1003.
    1. Gregoriadis G, Ryman BE. Liposomes as carriers of enzymes or drugs: a new approach to the treatment of storage diseases. Biochem J. 1971;124:58P.
    1. Dromi S, Frenkel V, Luk A. Pulsed-high intensity focused ultrasound and low temperature–sensitive liposomes for enhanced targeted drug delivery and antitumor effect. Clin Cancer Res. 2007;13:2722–2727.
    1. Needham D, Anyarambhatla G, Kong G, Dewhirst MW. A new temperature-sensitive liposome for use with mild hyperthermia: characterization and testing in a human tumor xenograft model. Cancer Res. 2000;60:1197–1201.
    1. Staruch RM, Ganguly M, Tannock IF, Hynynen K, Chopra R. Enhanced drug delivery in rabbit VX2 tumours using thermosensitive liposomes and MRI-controlled focused ultrasound hyperthermia. Int J Hyperth. 2012;28:776–787.
    1. Staruch RM, Hynynen K, Chopra R. Hyperthermia-mediated doxorubicin release from thermosensitive liposomes using MR-HIFU: therapeutic effect in rabbit Vx2 tumours. Int J Hyperth. 2015;31:118–133.
    1. Gabizon A, Shmeeda H, Barenholz Y. Pharmacokinetics of pegylated liposomal doxorubicin: review of animal and human studies. Clin Pharmacokinet. 2003;42:419–436.
    1. Wood BJ, Poon RT, Locklin JK. Phase I study of heat-deployed liposomal doxorubicin during radiofrequency ablation for hepatic malignancies. J Vasc Interv Radiol. 2012;23:248–255.
    1. Poon RT, Borys N. Lyso-thermosensitive liposomal doxorubicin: an adjuvant to increase the cure rate of radiofrequency ablation in liver cancer. Futur Oncol. 2011;7:937–945.
    1. Tak WY, Lin S-M, Wang Y. Phase III HEAT study adding lyso-thermosensitive liposomal doxorubicin to radiofrequency ablation in patients with unresectable hepatocellular carcinoma lesions. Clin Cancer Res. 2017;24:73–83.
    1. Lyon PC, Griffiths LF, Lee J. Clinical trial protocol for TARDOX: a phase I study to investigate the feasibility of targeted release of lyso-thermosensitive liposomal doxorubicin (ThermoDox®) using focused ultrasound in patients with liver tumours. J Ther Ultrasound. 2017;5:28.
    1. Choi H, Charnsangavej C, Faria SC. Correlation of computed tomography and positron emission tomography in patients with metastatic gastrointestinal stromal tumor treated at a single institution with imatinib mesylate: proposal of new computed tomography response criteria. J Clin Oncol. 2007;25:1753–1759.
    1. Eisenhauer EA, Therasse P, Bogaerts J. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1) Eur J Cancer. 2009;45:228–247.
    1. Wahl RL, Jacene H, Kasamon Y, Lodge MA. From RECIST to PERCIST: evolving considerations for PET response criteria in solid tumors. J Nucl Med. 2009;50(suppl 1):122S–150S.
    1. Larson SM, Erdi Y, Akhurst T. Tumor treatment response based on visual and quantitative changes in global tumor glycolysis using PET-FDG imaging. The visual response score and the change in total lesion glycolysis. Clin Positron Imaging. 1999;2:159–171.
    1. Hynynen K, Martin CJ, Watmough DJ, Mallard JR. Errors in temperature measurement by thermocouple probes during ultrasound induced hyperthermia. Br J Radiol. 1983;56:969–970.
    1. Vaezy S, Shi X, Martin RW. Real-time visualization of high-intensity focused ultrasound treatment using ultrasound imaging. Ultrasound Med Biol. 2001;27:33–42.
    1. Kreb DL, Bosscha K, Ernst MF. Use of cytokeratin 8 immunohistochemistry for assessing cell death after radiofrequency ablation of breast cancers. Biotech Histochem. 2011;86:404–412.
    1. Poon RT, Borys N. Lyso-thermosensitive liposomal doxorubicin: a novel approach to enhance efficacy of thermal ablation of liver cancer. Expert Opin Pharmacother. 2009;10:333–343.
    1. Laginha KM, Verwoert S, Charrois GJ, Allen TM. Determination of doxorubicin levels in whole tumor and tumor nuclei in murine breast cancer tumors. Clin Cancer Res. 2005;11:6944–6949.
    1. Maeda H. The enhanced permeability and retention (EPR) effect in tumor vasculature: the key role of tumor-selective macromolecular drug targeting. Adv Enzym Regul. 2001;41:189–207.
    1. Landon CD, Park J-Y, Needham D, Dewhirst MW. Nanoscale drug delivery and hyperthermia: the materials design and preclinical and clinical testing of low temperature-sensitive liposomes used in combination with mild hyperthermia in the treatment of local cancer. Open Nanomed J. 2011;3:38.
    1. Dawidczyk CM, Russell LM, Hultz M, Searson PC. Tumor accumulation of liposomal doxorubicin in three murine models: optimizing delivery efficiency. Nanomedicine. 2017;13:1637–1644.
    1. Kong G, Braun RD, Dewhirst MW. Hyperthermia enables tumor-specific nanoparticle delivery: effect of particle size. Cancer Res. 2000;60:4440–4445.
    1. Hobbs SK, Monsky WL, Yuan F. Regulation of transport pathways in tumor vessels: Role of tumor type and microenvironment. Proc Natl Acad Sci USA. 1998;95:4607–4612.
    1. Hahn GM, Braun J, Har-Kedar I. Thermochemotherapy: synergism between hyperthermia (42–43 degrees) and adriamycin (of bleomycin) in mammalian cell inactivation. Proc Natl Acad Sci USA. 1975;72:937–940.
    1. Myers R, Coviello C, Erbs P. Polymeric cups for cavitation-mediated delivery of oncolytic vaccinia virus. Mol Ther. 2016;24:1627–1633.
    1. Nwokeoha S, Carlisle R, Cleveland RO. The application of clinical lithotripter shock waves to RNA nucleotide delivery to cells. Ultrasound Med Biol. 2016;42:2478–2492.
    1. Welch DR. Tumor heterogeneity—a ‘contemporary concept’ founded on historical insights and predictions. Cancer Res. 2016;76:4–6.
    1. Carlisle R, Coussios CC. Mechanical approaches to oncological drug delivery. Ther Deliv. 2013;4:1213–1215.
    1. Staruch R, Chopra R, Hynynen K. Localised drug release using MRI-controlled focused ultrasound hyperthermia. Int J Hyperth. 2011;27:156–171.
    1. Hijnen N, Langereis S, Grüll H. Magnetic resonance guided high-intensity focused ultrasound for image-guided temperature-induced drug delivery. Adv Drug Deliv Rev. 2014;72:65–81.
    1. Weinländer G, Kornek G, Raderer M, Hejna M, Tetzner C, Scheithauer W. Treatment of advanced colorectal cancer with doxorubicin combined with two potential multidrug-resistance-reversing agents: high-dose oral tamoxifen and dexverapamil. J Cancer Res Clin Oncol. 1997;123:452–455.

Source: PubMed

3
Sottoscrivi