Prospective randomized trial of enoxaparin, pentoxifylline and ursodeoxycholic acid for prevention of radiation-induced liver toxicity

Max Seidensticker, Ricarda Seidensticker, Robert Damm, Konrad Mohnike, Maciej Pech, Bruno Sangro, Peter Hass, Peter Wust, Siegfried Kropf, Günther Gademann, Jens Ricke, Max Seidensticker, Ricarda Seidensticker, Robert Damm, Konrad Mohnike, Maciej Pech, Bruno Sangro, Peter Hass, Peter Wust, Siegfried Kropf, Günther Gademann, Jens Ricke

Abstract

Background/aim: Targeted radiotherapy of liver malignancies has found to be effective in selected patients. A key limiting factor of these therapies is the relatively low tolerance of the liver parenchyma to radiation. We sought to assess the preventive effects of a combined regimen of pentoxifylline (PTX), ursodeoxycholic acid (UDCA) and low-dose low molecular weight heparin (LMWH) on focal radiation-induced liver injury (fRILI).

Methods and materials: Patients with liver metastases from colorectal carcinoma who were scheduled for local ablation by radiotherapy (image-guided high-dose-rate interstitial brachytherapy) were prospectively randomized to receive PTX, UDCA and LMWH for 8 weeks (treatment) or no medication (control). Focal RILI at follow-up was assessed using functional hepatobiliary magnetic resonance imaging (MRI). A minimal threshold dose, i.e. the dose to which the outer rim of the fRILI was formerly exposed to, was quantified by merging MRI and dosimetry data.

Results: Results from an intended interim-analysis made a premature termination necessary. Twenty-two patients were included in the per-protocol analysis. Minimal mean hepatic threshold dose 6 weeks after radiotherapy (primary endpoint) was significantly higher in the study treatment-group compared with the control (19.1 Gy versus 14.6 Gy, p = 0.011). Qualitative evidence of fRILI by MRI at 6 weeks was observed in 45.5% of patients in the treatment versus 90.9% of the control group. No significant differences between the groups were observed at the 12-week follow-up.

Conclusions: The post-therapeutic application of PTX, UDCA and low-dose LMWH significantly reduced the extent and incidence fRILI at 6 weeks after radiotherapy. The development of subsequent fRILI at 12 weeks (4 weeks after cessation of PTX, UDCA and LMWH during weeks 1-8) in the treatment group was comparable to the control group thus supporting the observation that the agents mitigated fRILI.

Trial registration: EU clinical trials register 2008-002985-70 ClinicalTrials.gov NCT01149304.

Conflict of interest statement

Competing Interests: M. Seidensticker has served as a speaker for Bayer Healthcare and Sirtex medical, and has received research funding from Sirtex medical. R. Seidensticker has served as a speaker for Bayer Healthcare and Sirtex medical, and has received research funding from Sirtex medical. J. Ricke has served as a speaker for Bayer Healthcare and Sirtex medical, and has received research funding from Sirtex medical, Bayer Healthcare and Siemens. M. Pech has served as a speaker for Sirtex medical. B. Sangro has served as a speaker and an advisory board member for Sirtex medical. The authors state herewith that the competing interest as listed above do not alter their adherence to PLOS ONE policies on sharing data and materials.

Figures

Figure 1. T1w-axial THRIVE 20 min after…
Figure 1. T1w-axial THRIVE 20 min after application of Gd-EOB-DTPA (A, C–E and G, H) and BT planning CT with dosimetry (B and F).
A–D, control group. A: pre-treatment MRI displaying a metastasis scheduled for BT treatment (black arrow). B: Planning-CT after introduction of the brachytherapy catheters (black arrows). Clinical target volume (CTV) represented by bold red circle and dosimetry by coloured lines (red: 20 Gy-, blue: 12 Gy-isodose). C: MRI at 6 weeks showing substantial reduction in Gd-EOB-DTPA uptake by liver parenchyma adjacent to treated metastases (i.e. focal radiation-induced liver injury, fRILI). Note: The area of fRILI matches the geometry of the dosimetry (B). Determined threshold dose: 9.75 Gy. D: MRI at 3 months showing shrinkage of the fRILI. Determined threshold dose: 11.9 Gy. E–H, treatment group. E: pre-treatment MRI displaying two metastases (black arrow); two more treated lesions are not displayed in the plane. F: Planning-CT (annotations: see B). G: MRI at 6 weeks showing no fRILI. H: MRI at 3 months after radiotherapy (and 1 month after finishing study treatment) showing a substantial region of fRILI. Determined threshold dose: 15.8 Gy.
Figure 2. CONSORT-diagram.
Figure 2. CONSORT-diagram.
*Exclusion criterion age was initially disregarded by error in this patient (aged 82). **Exclusion criterion prior radiotherapy was initially disregarded by error in this patient (prior radiotherapy was performed 2 years earlier with location in the contralateral liver lobe).

References

    1. Boda-Heggemann J, Dinter D, Weiss C, Frauenfeld A, Siebenlist K, et al. (2012) Hypofractionated image-guided breath-hold SABR (stereotactic ablative body radiotherapy) of liver metastases–clinical results. Radiation oncology 7: 92.
    1. Ricke J, Mohnike K, Pech M, Seidensticker M, Ruhl R, et al. (2010) Local response and impact on survival after local ablation of liver metastases from colorectal carcinoma by computed tomography-guided high-dose-rate brachytherapy. International journal of radiation oncology, biology, physics 78: 479–485.
    1. Seidensticker R, Denecke T, Kraus P, Seidensticker M, Mohnike K, et al. (2012) Matched-pair comparison of radioembolization plus best supportive care versus best supportive care alone for chemotherapy refractory liver-dominant colorectal metastases. Cardiovascular and interventional radiology 35: 1066–1073.
    1. Emami B, Lyman J, Brown A, Coia L, Goitein M, et al. (1991) Tolerance of normal tissue to therapeutic irradiation. International journal of radiation oncology, biology, physics 21: 109–122.
    1. Lawrence TS, Robertson JM, Anscher MS, Jirtle RL, Ensminger WD, et al. (1995) Hepatic toxicity resulting from cancer treatment. International journal of radiation oncology, biology, physics 31: 1237–1248.
    1. Ricke J, Seidensticker M, Ludemann L, Pech M, Wieners G, et al. (2005) In vivo assessment of the tolerance dose of small liver volumes after single-fraction HDR irradiation. International journal of radiation oncology, biology, physics 62: 776–784.
    1. Seidensticker M, Seidensticker R, Mohnike K, Wybranski C, Kalinski T, et al. (2011) Quantitative in vivo assessment of radiation injury of the liver using Gd-EOB-DTPA enhanced MRI: tolerance dose of small liver volumes. Radiation oncology 6: 40.
    1. McDonald GB, Sharma P, Matthews DE, Shulman HM, Thomas ED (1985) The clinical course of 53 patients with venocclusive disease of the liver after marrow transplantation. Transplantation 39: 603–608.
    1. Sangro B, Gil-Alzugaray B, Rodriguez J, Sola I, Martinez-Cuesta A, et al. (2008) Liver disease induced by radioembolization of liver tumors: description and possible risk factors. Cancer 112: 1538–1546.
    1. Farthing MJ, Clark ML, Sloane JP, Powles RL, McElwain TJ (1982) Liver disease after bone marrow transplantation. Gut 23: 465–474.
    1. Fajardo LF, Colby TV (1980) Pathogenesis of veno-occlusive liver disease after radiation. Archives of pathology & laboratory medicine 104: 584–588.
    1. Reed GB Jr, Cox AJ Jr (1966) The human liver after radiation injury. A form of veno-occlusive disease. The American journal of pathology 48: 597–611.
    1. Shulman HM, Gown AM, Nugent DJ (1987) Hepatic veno-occlusive disease after bone marrow transplantation. Immunohistochemical identification of the material within occluded central venules. The American journal of pathology 127: 549–558.
    1. Attal M, Huguet F, Rubie H, Charlet JP, Schlaifer D, et al. (1993) Prevention of regimen-related toxicities after bone marrow transplantation by pentoxifylline: a prospective, randomized trial. Blood 82: 732–736.
    1. Attal M, Huguet F, Rubie H, Huynh A, Charlet JP, et al. (1992) Prevention of hepatic veno-occlusive disease after bone marrow transplantation by continuous infusion of low-dose heparin: a prospective, randomized trial. Blood 79: 2834–2840.
    1. Bianco JA, Appelbaum FR, Nemunaitis J, Almgren J, Andrews F, et al. (1991) Phase I-II trial of pentoxifylline for the prevention of transplant-related toxicities following bone marrow transplantation. Blood 78: 1205–1211.
    1. Essell JH, Schroeder MT, Harman GS, Halvorson R, Lew V, et al. (1998) Ursodiol prophylaxis against hepatic complications of allogeneic bone marrow transplantation. A randomized, double-blind, placebo-controlled trial. Annals of internal medicine 128: 975–981.
    1. Forrest DL, Thompson K, Dorcas VG, Couban SH, Pierce R (2003) Low molecular weight heparin for the prevention of hepatic veno-occlusive disease (VOD) after hematopoietic stem cell transplantation: a prospective phase II study. Bone marrow transplantation 31: 1143–1149.
    1. Ohashi K, Tanabe J, Watanabe R, Tanaka T, Sakamaki H, et al. (2000) The Japanese multicenter open randomized trial of ursodeoxycholic acid prophylaxis for hepatic veno-occlusive disease after stem cell transplantation. American journal of hematology 64: 32–38.
    1. Or R, Nagler A, Shpilberg O, Elad S, Naparstek E, et al. (1996) Low molecular weight heparin for the prevention of veno-occlusive disease of the liver in bone marrow transplantation patients. Transplantation 61: 1067–1071.
    1. Ruutu T, Eriksson B, Remes K, Juvonen E, Volin L, et al. (2002) Ursodeoxycholic acid for the prevention of hepatic complications in allogeneic stem cell transplantation. Blood 100: 1977–1983.
    1. Clift RA, Bianco JA, Appelbaum FR, Buckner CD, Singer JW, et al. (1993) A randomized controlled trial of pentoxifylline for the prevention of regimen-related toxicities in patients undergoing allogeneic marrow transplantation. Blood 82: 2025–2030.
    1. McDonald GB, Sharma P, Matthews DE, Shulman HM, Thomas ED (1984) Venocclusive disease of the liver after bone marrow transplantation: diagnosis, incidence, and predisposing factors. Hepatology 4: 116–122.
    1. Seidensticker M, Burak M, Kalinski T, Garlipp B, Koelble K, et al. (2014) Radiation-Induced Liver Damage: Correlation of Histopathology with Hepatobiliary Magnetic Resonance Imaging, a Feasibility Study. Cardiovascular and interventional radiology.
    1. Wybranski C, Seidensticker M, Mohnike K, Kropf S, Wust P, et al. (2009) In vivo assessment of dose volume and dose gradient effects on the tolerance dose of small liver volumes after single-fraction high-dose-rate 192Ir irradiation. Radiation research 172: 598–606.
    1. Shulman HM, Hinterberger W (1992) Hepatic veno-occlusive disease–liver toxicity syndrome after bone marrow transplantation. Bone marrow transplantation 10: 197–214.
    1. Lakshminarayanan S, Sahdev I, Goyal M, Vlachos A, Atlas M, et al. (2010) Low incidence of hepatic veno-occlusive disease in pediatric patients undergoing hematopoietic stem cell transplantation attributed to a combination of intravenous heparin, oral glutamine, and ursodiol at a single transplant institution. Pediatric transplantation 14: 618–621.
    1. Park SH, Lee MH, Lee H, Kim HS, Kim K, et al. (2002) A randomized trial of heparin plus ursodiol vs. heparin alone to prevent hepatic veno-occlusive disease after hematopoietic stem cell transplantation. Bone marrow transplantation 29: 137–143.
    1. Lee JH, Lee KH, Kim S, Lee JS, Kim WK, et al. (1998) Relevance of proteins C and S, antithrombin III, von Willebrand factor, and factor VIII for the development of hepatic veno-occlusive disease in patients undergoing allogeneic bone marrow transplantation: a prospective study. Bone marrow transplantation 22: 883–888.
    1. Catani L, Gugliotta L, Vianelli N, Nocentini F, Baravelli S, et al. (1996) Endothelium and bone marrow transplantation. Bone marrow transplantation 17: 277–280.
    1. Geraci JP, Mariano MS (1993) Radiation hepatology of the rat: parenchymal and nonparenchymal cell injury. Radiation research 136: 205–213.
    1. Kowdley KV (2000) Ursodeoxycholic acid therapy in hepatobiliary disease. The American journal of medicine 108: 481–486.
    1. Neuman MG, Shear NH, Bellentani S, Tiribelli C (1998) Role of cytokines in ethanol-induced cytotoxicity in vitro in Hep G2 cells. Gastroenterology 115: 157–166.
    1. Lindner H, Holler E, Ertl B, Multhoff G, Schreglmann M, et al. (1997) Peripheral blood mononuclear cells induce programmed cell death in human endothelial cells and may prevent repair: role of cytokines. Blood 89: 1931–1938.
    1. Rodrigues CM, Fan G, Ma X, Kren BT, Steer CJ (1998) A novel role for ursodeoxycholic acid in inhibiting apoptosis by modulating mitochondrial membrane perturbation. The Journal of clinical investigation 101: 2790–2799.
    1. Pascolo L, Cupelli F, Anelli PL, Lorusso V, Visigalli M, et al. (1999) Molecular mechanisms for the hepatic uptake of magnetic resonance imaging contrast agents. Biochemical and biophysical research communications 257: 746–752.
    1. Schuhmann-Giampieri G, Schmitt-Willich H, Press WR, Negishi C, Weinmann HJ, et al. (1992) Preclinical evaluation of Gd-EOB-DTPA as a contrast agent in MR imaging of the hepatobiliary system. Radiology 183: 59–64.
    1. Watanabe H, Kanematsu M, Goshima S, Kondo H, Onozuka M, et al. (2011) Staging hepatic fibrosis: comparison of gadoxetate disodium-enhanced and diffusion-weighted MR imaging–preliminary observations. Radiology 259: 142–150.
    1. Shin NY, Kim MJ, Lim JS, Park MS, Chung YE, et al. (2012) Accuracy of gadoxetic acid-enhanced magnetic resonance imaging for the diagnosis of sinusoidal obstruction syndrome in patients with chemotherapy-treated colorectal liver metastases. European radiology 22: 864–871.

Source: PubMed

3
订阅