Hyperthermic Laser Ablation of Recurrent Glioblastoma Leads to Temporary Disruption of the Peritumoral Blood Brain Barrier

Eric C Leuthardt, Chong Duan, Michael J Kim, Jian L Campian, Albert H Kim, Michelle M Miller-Thomas, Joshua S Shimony, David D Tran, Eric C Leuthardt, Chong Duan, Michael J Kim, Jian L Campian, Albert H Kim, Michelle M Miller-Thomas, Joshua S Shimony, David D Tran

Abstract

Background: Poor central nervous system penetration of cytotoxic drugs due to the blood brain barrier (BBB) is a major limiting factor in the treatment of brain tumors. Most recurrent glioblastomas (GBM) occur within the peritumoral region. In this study, we describe a hyperthemic method to induce temporary disruption of the peritumoral BBB that can potentially be used to enhance drug delivery.

Methods: Twenty patients with probable recurrent GBM were enrolled in this study. Fourteen patients were evaluable. MRI-guided laser interstitial thermal therapy was applied to achieve both tumor cytoreduction and disruption of the peritumoral BBB. To determine the degree and timing of peritumoral BBB disruption, dynamic contrast-enhancement brain MRI was used to calculate the vascular transfer constant (Ktrans) in the peritumoral region as direct measures of BBB permeability before and after laser ablation. Serum levels of brain-specific enolase, also known as neuron-specific enolase, were also measured and used as an independent quantification of BBB disruption.

Results: In all 14 evaluable patients, Ktrans levels peaked immediately post laser ablation, followed by a gradual decline over the following 4 weeks. Serum BSE concentrations increased shortly after laser ablation and peaked in 1-3 weeks before decreasing to baseline by 6 weeks.

Conclusions: The data from our pilot research support that disruption of the peritumoral BBB was induced by hyperthemia with the peak of high permeability occurring within 1-2 weeks after laser ablation and resolving by 4-6 weeks. This provides a therapeutic window of opportunity during which delivery of BBB-impermeant therapeutic agents may be enhanced.

Trial registration: ClinicalTrials.gov NCT01851733.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Fig 1. CONSORT flow diagram of the…
Fig 1. CONSORT flow diagram of the BBB disruption measurement portion of the pilot phase 2 study involving the first 20 enrolled patients.
Early or Early doxorubicin: Treatment started within 1 week after LITT. Late or Late doxorubicin: Treatment started at 6 weeks after LITT.
Fig 2. Radiographic appearances of post-LITT changes.
Fig 2. Radiographic appearances of post-LITT changes.
(A) A woman with a left thalamic GBM treated with LITT underwent axial and coronal T1-weighted post-contrast enhanced MR images of the brain pre-LITT, during LITT and 48 hours post LITT. (B-I) A woman with a left insula GBM underwent axial T1-weighted post-contrast enhanced (B-E) and axial FLAIR-weighted (F-I) MR images of the brain pre-LITT and within 48 hours post LITT, 2 weeks post LITT, and 4 weeks post LITT. In both cases, the enhancing tumor (solid red arrowheads in A and B) is replaced by a central zone of T1-weighted signal hyperintensity (open red arrowheads in A and D) and a faint, new discontinuous rim of enhancement extending beyond the original tumor associated enhancing rim (solid blue arrowheads in A and D). The rim of contrast enhancement intensifies at 2 weeks post LITT (D) and remains stable at 4 weeks post LITT (E). Perilesional edema evaluated on FLAIR-weighted images is slightly increased between the pre-treatment (F) and immediate post-treatment (G) images, increases to a maximum point on the 2-week post-treatment images (H) and improves slightly by the 4-week images (I). The orange circles denote a representative ROI used to calculate temporal progression of Ktrans after LITT
Fig 3. Peritumoral BBB disruption induced by…
Fig 3. Peritumoral BBB disruption induced by LITT as measured by DCE-MRI.
Ktrans for each of the 14 eligible subjects in the study as a function of time in days from the LITT procedure. In all subjects the Ktrans is highly elevated in the first few days after the procedure and then progressively decreases out to approximately the 4-week time point. This is best illustrated in the bottom right blue graph, which is an average of the 14 subject curves.
Fig 4. Optimization of the BSE ELISA…
Fig 4. Optimization of the BSE ELISA assay for measuring BBB disruption.
Serum concentrations of BSE before and after open craniotomy for surgical debulking in 3 subjects (A, B, and C) with a low-grade glioma, WHO grade II. *p

Fig 5. BBB disruption induced by LITT…

Fig 5. BBB disruption induced by LITT as measured by serum biomarkers.

Serum concentrations of…

Fig 5. BBB disruption induced by LITT as measured by serum biomarkers.
Serum concentrations of BSE for each of the 14 evaluable subjects in the study (A-N) and as the mean + SEM (O) as a function of time in days from the LITT procedure. In 7/14 subjects, serum BSE levels slightly decreased immediately after LITT, then in 13/14 subjects, serum BSE levels rose shortly after LITT, peaked between 1–3 weeks after LITT, and then decreased by the 6-week time point. In Patient #12, serum BSE concentration increased at week 10 coincident with an increased Ktrans at the same time point, consistent with a recurrent tumor as demonstrated on diagnostic MR imaging. Patient #15’s serum BSE concentration began to rise by week 4, consistent with early multifocal recurrent disease as demonstrated on diagnostic MR imaging.
Fig 5. BBB disruption induced by LITT…
Fig 5. BBB disruption induced by LITT as measured by serum biomarkers.
Serum concentrations of BSE for each of the 14 evaluable subjects in the study (A-N) and as the mean + SEM (O) as a function of time in days from the LITT procedure. In 7/14 subjects, serum BSE levels slightly decreased immediately after LITT, then in 13/14 subjects, serum BSE levels rose shortly after LITT, peaked between 1–3 weeks after LITT, and then decreased by the 6-week time point. In Patient #12, serum BSE concentration increased at week 10 coincident with an increased Ktrans at the same time point, consistent with a recurrent tumor as demonstrated on diagnostic MR imaging. Patient #15’s serum BSE concentration began to rise by week 4, consistent with early multifocal recurrent disease as demonstrated on diagnostic MR imaging.

References

    1. Parkin DM. Global cancer statistics in the year 2000. The Lancet Oncology. 2001;2(9):533–43.
    1. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJB, et al. Radiotherapy plus Concomitant and Adjuvant Temozolomide for Glioblastoma. New England Journal of Medicine. 2005;352(10):987–96. 10.1056/NEJMoa043330
    1. Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJB, Janzer RC, et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. The Lancet Oncology. 2009;10(5):459–66. 10.1016/S1470-2045(09)70025-7
    1. Hochberg FH, Pruitt A. Assumptions in the radiotherapy of glioblastoma. Neurology. 1980;30(9):907–11. Epub 1980/09/01. .
    1. Lemée J-M, Clavreul A, Menei P. Intratumoral heterogeneity in glioblastoma: don't forget the peritumoral brain zone. Neuro-Oncology. 2015;17(10):1322–32. 10.1093/neuonc/nov119
    1. Holodny AI, Nusbaum AO, Festa S, Pronin IN, Lee HJ, Kalnin AJ. Correlation between the degree of contrast enhancement and the volume of peritumoral edema in meningiomas and malignant gliomas. Neuroradiology. 1999;41(11):820–5. Epub 1999/12/22. .
    1. Kassner A, Thornhill R. Measuring the integrity of the human blood-brain barrier using magnetic resonance imaging. Methods Mol Biol. 2011;686:229–45. Epub 2010/11/18. 10.1007/978-1-60761-938-3_10 .
    1. Deeken JF, L√∂scher W. The Blood-Brain Barrier and Cancer: Transporters, Treatment, and Trojan Horses. Clinical Cancer Research. 2007;13(6):1663–74. 10.1158/1078-0432.ccr-06-2854
    1. Stewart DJ, Richard MT, Hugenholtz H, Dennery JM, Belanger R, Gerin-Lajoie J, et al. Penetration of VP-16 (etoposide) into human intracerebral and extracerebral tumors. J Neurooncol. 1984;2(2):133–9. Epub 1984/01/01. .
    1. Stewart DJ, Lu K, Benjamin RS, Leavens ME, Luna M, Yap HY, et al. Concentration of vinblastine in human intracerebral tumor and other tissues. J Neurooncol. 1983;1(2):139–44. Epub 1983/01/01. 6678966.
    1. Brem H, Piantadosi S, Burger PC, Walker M, Selker R, Vick NA, et al. Placebo-controlled trial of safety and efficacy of intraoperative controlled delivery by biodegradable polymers of chemotherapy for recurrent gliomas. The Polymer-brain Tumor Treatment Group. Lancet. 1995;345(8956):1008–12. Epub 1995/04/22. .
    1. Westphal M, Ram Z, Riddle V, Hilt D, Bortey E. Gliadel wafer in initial surgery for malignant glioma: long-term follow-up of a multicenter controlled trial. Acta Neurochir (Wien). 2006;148(3):269–75; discussion 75. Epub 2006/02/17. 10.1007/s00701-005-0707-z .
    1. Westphal M, Hilt DC, Bortey E, Delavault P, Olivares R, Warnke PC, et al. A phase 3 trial of local chemotherapy with biodegradable carmustine (BCNU) wafers (Gliadel wafers) in patients with primary malignant glioma. Neuro-oncology. 2003;5(2):79–88. Epub 2003/04/04. 10.1215/S1522-8517-02-00023-6
    1. Fleming AB, Saltzman WM. Pharmacokinetics of the carmustine implant. Clin Pharmacokinet. 2002;41(6):403–19. Epub 2002/06/21. .
    1. Bobo RH, Laske DW, Akbasak A, Morrison PF, Dedrick RL, Oldfield EH. Convection-enhanced delivery of macromolecules in the brain. Proceedings of the National Academy of Sciences. 1994;91(6):2076–80.
    1. Gong W, Wang Z, Liu N, Lin W, Wang X, Xu D, et al. Improving efficiency of adriamycin crossing blood brain barrier by combination of thermosensitive liposomes and hyperthermia. Biol Pharm Bull. 2011;34(7):1058–64. Epub 2011/07/02. .
    1. Oztas B, Kucuk M. Reversible blood-brain barrier dysfunction after intracarotid hyperthermic saline infusion. Int J Hyperthermia. 1998;14(4):395–401. Epub 1998/08/05. .
    1. Sabel M, Rommel F, Kondakci M, Gorol M, Willers R, Bilzer T. Locoregional opening of the rodent blood-brain barrier for paclitaxel using Nd:YAG laser-induced thermo therapy: a new concept of adjuvant glioma therapy? Lasers Surg Med. 2003;33(2):75–80. Epub 2003/08/13. 10.1002/lsm.10181 .
    1. Kangasniemi M, McNichols RJ, Bankson JA, Gowda A, Price RE, Hazle JD. Thermal therapy of canine cerebral tumors using a 980 nm diode laser with MR temperature-sensitive imaging feedback. Lasers Surg Med. 2004;35(1):41–50. 10.1002/lsm.20069 .
    1. Reimer P, Bremer C, Horch C, Morgenroth C, Allkemper T, Schuierer G. MR-monitored LITT as a palliative concept in patients with high grade gliomas: preliminary clinical experience. J Magn Reson Imaging. 1998;8(1):240–4. .
    1. Schulze PC, Kahn T, Harth T, Schwurzmaier HJ, Schober R. Correlation of neuropathologic findings and phase-based MRI temperature maps in experimental laser-induced interstitial thermotherapy. J Magn Reson Imaging. 1998;8(1):115–20. .
    1. Schwabe B, Kahn T, Harth T, Ulrich F, Schwarzmaier HJ. Laser-induced thermal lesions in the human brain: short- and long-term appearance on MRI. J Comput Assist Tomogr. 1997;21(5):818–25. .
    1. Kangasniemi M, Stafford RJ, Price RE, Jackson EF, Hazle JD. Dynamic gadolinium uptake in thermally treated canine brain tissue and experimental cerebral tumors. Invest Radiol. 2003;38(2):102–7. 10.1097/01.RLI.0000044933.76182.49 .
    1. Hawasli AH, Kim AH, Dunn GP, Tran DD, Leuthardt EC. Stereotactic laser ablation of high-grade gliomas. Neurosurg Focus. 2014;37(6):E1 10.3171/2014.9.FOCUS14471 .
    1. Mohammadi AM, Hawasli AH, Rodriguez A, Schroeder JL, Laxton AW, Elson P, et al. The role of laser interstitial thermal therapy in enhancing progression-free survival of difficult-to-access high-grade gliomas: a multicenter study. Cancer Med. 2014;3(4):971–9. 10.1002/cam4.266
    1. Hawasli AH, Bagade S, Shimony JS, Miller-Thomas M, Leuthardt EC. Magnetic resonance imaging-guided focused laser interstitial thermal therapy for intracranial lesions: single-institution series. Neurosurgery. 2013;73(6):1007–17. 10.1227/NEU.0000000000000144
    1. Hawasli AH, Ray WZ, Murphy RK, Dacey RG Jr., Leuthardt EC. Magnetic resonance imaging-guided focused laser interstitial thermal therapy for subinsular metastatic adenocarcinoma: technical case report. Neurosurgery. 2012;70(2 Suppl Operative):332–7; discussion 8. Epub 2011/08/27. 10.1227/NEU.0b013e318232fc90 .
    1. Tofts PS, Brix G, Buckley DL, Evelhoch JL, Henderson E, Knopp MV, et al. Estimating kinetic parameters from dynamic contrast-enhanced T(1)-weighted MRI of a diffusable tracer: standardized quantities and symbols. J Magn Reson Imaging. 1999;10(3):223–32. .
    1. Leach MO, Morgan B, Tofts PS, Buckley DL, Huang W, Horsfield MA, et al. Imaging vascular function for early stage clinical trials using dynamic contrast-enhanced magnetic resonance imaging. Eur Radiol. 2012;22(7):1451–64. 10.1007/s00330-012-2446-x .
    1. Kallehauge J, Nielsen T, Haack S, Peters DA, Mohamed S, Fokdal L, et al. Voxelwise comparison of perfusion parameters estimated using dynamic contrast enhanced (DCE) computed tomography and DCE-magnetic resonance imaging in locally advanced cervical cancer. Acta Oncol. 2013;52(7):1360–8. 10.3109/0284186X.2013.813637 .
    1. Sommer JC, Schmid VJ. Spatial two-tissue compartment model for dynamic contrast-enhanced magnetic resonance imaging. Journal of the Royal Statistical Society: Series C (Applied Statistics). 2014;63(5):695–713. 10.1111/rssc.12057
    1. Zhou J, Wilson DA, Ulatowski JA, Traystman RJ, van Zijl PCM. Two-Compartment Exchange Model for Perfusion Quantification Using Arterial Spin Tagging. J Cereb Blood Flow Metab. 2001;21(4):440–55.
    1. Donaldson SB, West CM, Davidson SE, Carrington BM, Hutchison G, Jones AP, et al. A comparison of tracer kinetic models for T1-weighted dynamic contrast-enhanced MRI: application in carcinoma of the cervix. Magn Reson Med. 2010;63(3):691–700. 10.1002/mrm.22217 .
    1. Lee JJ, Xuemin G, Suyu L. Bayesian adaptive randomization designs for targeted agent development. Clin Trials. 2010;7(5):584–96. 10.1177/1740774510373120 .
    1. Correale J, Rabinowicz AL, Heck CN, Smith TD, Loskota WJ, DeGiorgio CM. Status epilepticus increases CSF levels of neuron-specific enolase and alters the blood-brain barrier. Neurology. 1998;50(5):1388–91. Epub 1998/05/22. .
    1. Marchi N, Rasmussen P, Kapural M, Fazio V, Kight K, Mayberg MR, et al. Peripheral markers of brain damage and blood-brain barrier dysfunction. Restor Neurol Neurosci. 2003;21(3–4):109–21. Epub 2003/10/08. .
    1. Stalnacke BM, Tegner Y, Sojka P. Playing soccer increases serum concentrations of the biochemical markers of brain damage S-100B and neuron-specific enolase in elite players: a pilot study. Brain Inj. 2004;18(9):899–909. 10.1080/02699050410001671865 .
    1. Mondello S, Muller U, Jeromin A, Streeter J, Hayes RL, Wang KK. Blood-based diagnostics of traumatic brain injuries. Expert Rev Mol Diagn. 2011;11(1):65–78. 10.1586/erm.10.104
    1. Schaarschmidt H, Prange HW, Reiber H. Neuron-specific enolase concentrations in blood as a prognostic parameter in cerebrovascular diseases. Stroke. 1994;25(3):558–65. .

Source: PubMed

3
Abonneren