Modeling of Normal Tissue Complications Using Imaging and Biomarkers After Radiation Therapy for Hepatocellular Carcinoma.

PURPOSE To develop normal tissue complications (NTCP) models for hepatocellular cancer (HCC) patients who undergo liver radiation therapy (RT) and to evaluate the potential role of functional imaging and measurement of blood-based circulating biological markers before and during RT to improve the performance of these models. METHODS AND MATERIALS The data from 192 HCC patients who had undergone RT from 2005 to 2014 were evaluated. Of the 192 patients, 146 had received stereotactic body RT (SBRT) and 46 had received conventional RT to a median physical tumor dose of 49.8 Gy and 50.4 Gy, respectively. The physical doses were converted into 2-Gy equivalents for analysis. Two approaches were investigated for modeling NTCP: (1) a generalized Lyman-Kutcher-Burman model; and (2) a generalization of the parallel architecture model. Three clinical endpoints were considered: the change in albumin-bilirubin (ALBI), change in Child-Pugh (C-P) score, and grade ≥3 liver enzymatic changes. Local dynamic contrast-enhanced magnetic resonance imaging portal venous perfusion information was used as an imaging biomarker for local liver function. Four candidate inflammatory cytokines were considered as biological markers. The imaging findings and cytokine levels were incorporated into NTCP modeling, and their role was evaluated using goodness-of-fit metrics. RESULTS Using dosimetric information only, the Lyman-Kutcher-Burman model for the ALBI/C-P change had a steeper response curve compared with grade ≥3 enzymatic changes. Incorporating portal venous perfusion imaging information into the parallel architecture model to represent functional reserve resulted in relatively steeper dose-response curves compared with dose-only models. A larger loss of perfusion function was needed for enzymatic changes compared with ALBI/C-P changes. Increased transforming growth factor-β1 and eotaxin expression increased the trend of expected risk in both NTCP modeling approaches but did not reach statistical significance. CONCLUSIONS The incorporation of imaging findings and biological markers into NTCP modeling of liver toxicity improved the estimates of expected NTCP risk compared with using dose-only models. In addition, such generalized NTCP models should contribute to a better understanding of the normal tissue response in HCC SBRT patients and facilitate personalized treatment.

[1]  J. Sung,et al.  Biologic susceptibility of hepatocellular carcinoma patients treated with radiotherapy to radiation-induced liver disease. , 2004, International journal of radiation oncology, biology, physics.

[2]  Claudio Fiorino,et al.  Inclusion of clinical risk factors into NTCP modelling of late rectal toxicity after high dose radiotherapy for prostate cancer. , 2011, Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology.

[3]  M. Goitein,et al.  Fitting of normal tissue tolerance data to an analytic function. , 1991, International journal of radiation oncology, biology, physics.

[4]  J. Balter,et al.  Stereotactic body radiation therapy for primary and metastatic liver tumors. , 2013, Translational oncology.

[5]  S. Perkins,et al.  Phase I feasibility trial of stereotactic body radiation therapy for primary hepatocellular carcinoma , 2010, Clinical & translational oncology : official publication of the Federation of Spanish Oncology Societies and of the National Cancer Institute of Mexico.

[6]  Adam Johansson,et al.  An Overdetermined System of Transform Equations in Support of Robust DCE-MRI Registration With Outlier Rejection , 2016, Tomography.

[7]  H Wang,et al.  GPU-accelerated voxelwise hepatic perfusion quantification. , 2012, Physics in medicine and biology.

[8]  B. Kavanagh,et al.  Hepatic radiation toxicity: avoidance and amelioration. , 2011, Seminars in radiation oncology.

[9]  S. H. Cheng,et al.  Radiation-induced liver disease after radiotherapy for hepatocellular carcinoma: clinical manifestation and dosimetric description. , 2002, Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology.

[10]  Radhe Mohan,et al.  Incorporating single-nucleotide polymorphisms into the Lyman model to improve prediction of radiation pneumonitis. , 2013, International journal of radiation oncology, biology, physics.

[11]  N. Otsu A threshold selection method from gray level histograms , 1979 .

[12]  Andrea Bezjak,et al.  Stereotactic body radiation therapy for inoperable early stage lung cancer. , 2010, JAMA.

[13]  A. Agresti,et al.  Approximate is Better than “Exact” for Interval Estimation of Binomial Proportions , 1998 .

[14]  R. T. Ten Haken,et al.  Radiation-associated liver injury. , 2010, International journal of radiation oncology, biology, physics.

[15]  P. McCullagh,et al.  Generalized Linear Models , 1984 .

[16]  Joos V Lebesque,et al.  The benefits of including clinical factors in rectal normal tissue complication probability modeling after radiotherapy for prostate cancer. , 2012, International journal of radiation oncology, biology, physics.

[17]  G. Lockwood,et al.  Phase I study of individualized stereotactic body radiotherapy of liver metastases. , 2009, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[18]  M. Anscher,et al.  Transforming growth factor-beta 1 expression in irradiated liver. , 1990, Radiation research.

[19]  Cynthia S. Johnson,et al.  Stereotactic body radiotherapy for primary hepatocellular carcinoma. , 2011, International journal of radiation oncology, biology, physics.

[20]  R. Timmerman,et al.  Emergence of stereotactic body radiation therapy and its impact on current and future clinical practice. , 2014, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[21]  Daniel Normolle,et al.  Prediction of liver function by using magnetic resonance-based portal venous perfusion imaging. , 2013, International journal of radiation oncology, biology, physics.

[22]  Peter Balter,et al.  Stereotactic ablative radiotherapy versus lobectomy for operable stage I non-small-cell lung cancer: a pooled analysis of two randomised trials. , 2015, The Lancet. Oncology.

[23]  W. McBride,et al.  Cytokines in Radiobiological Responses: A Review , 2012, Radiation research.

[24]  R. Ahuja,et al.  Optimizing global liver function in radiation therapy treatment planning , 2016, Physics in medicine and biology.

[25]  K. Frey,et al.  Predictive models for regional hepatic function based on 99mTc-IDA SPECT and local radiation dose for physiologic adaptive radiation therapy. , 2013, International journal of radiation oncology, biology, physics.

[26]  J. Seong,et al.  Radiotherapeutic parameters predictive of liver complications induced by liver tumor radiotherapy. , 2009, International journal of radiation oncology, biology, physics.

[27]  M. Makuuchi,et al.  Assessment of hepatic reserve for indication of hepatic resection: decision tree incorporating indocyanine green test. , 2005, Journal of hepato-biliary-pancreatic surgery.

[28]  Yue Cao,et al.  Local and Global Function Model of the Liver. , 2016, International journal of radiation oncology, biology, physics.

[29]  S. Thorgeirsson,et al.  Role of growth hormone (GH) in liver regeneration. , 2004, Endocrinology.

[30]  A. Koong,et al.  Dose-escalation study of single-fraction stereotactic body radiotherapy for liver malignancies. , 2010, International journal of radiation oncology, biology, physics.

[31]  T. Tsuda,et al.  Comparison of Tc-99m-GSA scintigraphy with hepatic fibrosis and regeneration in patients with hepatectomy , 2003, Annals of nuclear medicine.

[32]  Daniel Normolle,et al.  Analysis of radiation-induced liver disease using the Lyman NTCP model. , 2002, International journal of radiation oncology, biology, physics.

[33]  S. Shiraishi,et al.  A quantitative index measured on 99mTc GSA SPECT/CT 3D fused images to evaluate severe fibrosis in patients with chronic liver disease , 2012, Japanese Journal of Radiology.

[34]  C. O’Farrelly,et al.  Liver immunology and its role in inflammation and homeostasis , 2016, Cellular & Molecular Immunology.

[35]  Yue Cao,et al.  Estimating functional liver reserve following hepatic irradiation: adaptive normal tissue response models. , 2014, Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology.

[36]  G J Kutcher,et al.  Analysis of clinical complication data for radiation hepatitis using a parallel architecture model. , 1995, International journal of radiation oncology, biology, physics.

[37]  I. E. Naqa,et al.  Radiation Sensitivity of the Liver: Models and Clinical Data , 2017 .

[38]  F. Tacke,et al.  Up‐regulated eotaxin plasma levels in chronic liver disease patients indicate hepatic inflammation, advanced fibrosis and adverse clinical course , 2007, Journal of gastroenterology and hepatology.

[39]  Albert C. Koong,et al.  Phase 2 Multi-institutional Trial Evaluating Gemcitabine and Stereotactic Body Radiotherapy for Patients With Locally Advanced Unresectable Pancreatic Adenocarcinoma , 2014, Cancer.

[40]  Daniel Normolle,et al.  Liver function after irradiation based on computed tomographic portal vein perfusion imaging. , 2008, International journal of radiation oncology, biology, physics.

[41]  H. Jang,et al.  Stereotactic body radiotherapy for patients with unresectable primary hepatocellular carcinoma: dose-volumetric parameters predicting the hepatic complication. , 2010, International journal of radiation oncology, biology, physics.

[42]  Xiao-dong Zhu,et al.  Prediction of radiation-induced liver disease by Lyman normal-tissue complication probability model in three-dimensional conformal radiation therapy for primary liver carcinoma. , 2006, International journal of radiation oncology, biology, physics.

[43]  Issam El Naqa,et al.  Contrasting analytical and data-driven frameworks for radiogenomic modeling of normal tissue toxicities in prostate cancer. , 2015, Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology.

[44]  P. Rubin,et al.  Blockade of CD40-CD40 ligand interactions protects against radiation-induced pulmonary inflammation and fibrosis. , 1998, Clinical immunology and immunopathology.

[45]  Zahra Kassam,et al.  Sequential phase I and II trials of stereotactic body radiotherapy for locally advanced hepatocellular carcinoma. , 2013, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[46]  Robert D Timmerman,et al.  A phase I trial of stereotactic body radiation therapy (SBRT) for liver metastases. , 2005, International journal of radiation oncology, biology, physics.

[47]  L. Dawson,et al.  Predictors of Liver Toxicity Following Stereotactic Body Radiation Therapy for Hepatocellular Carcinoma. , 2015, International journal of radiation oncology, biology, physics.

[48]  B. Sangro,et al.  Assessment of liver function in patients with hepatocellular carcinoma: a new evidence-based approach-the ALBI grade. , 2015, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[49]  M. Astrahan,et al.  Some implications of linear-quadratic-linear radiation dose-response with regard to hypofractionation. , 2008, Medical physics.