Review Open Access
Copyright ©The Author(s) 2018. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Jul 28, 2018; 24(28): 3090-3100
Published online Jul 28, 2018. doi: 10.3748/wjg.v24.i28.3090
Proton therapy for hepatocellular carcinoma: Current knowledges and future perspectives
Gyu Sang Yoo, Jeong Il Yu, Hee Chul Park, Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea
Hee Chul Park, Department of Medical Device Management and Research, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, South Korea
ORCID number: Gyu Sang Yoo (0000-0002-5542-5263); Jeong Il Yu (0000-0002-2009-7263); Hee Chul Park (0000-0003-0385-9681).
Author contributions: Yoo GS, Yu JI and Park HC contributed equally to this manuscript, including design of the study and literature review; Park HC supervised the project.
Supported by a Grant from the Marine Biotechnology Program funded by the Ministry of Oceans and Fisheries, South Korea, No. 20150220.
Conflict-of-interest statement: No potential conflicts of interest.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Hee Chul Park, MD, PhD, Professor, Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, South Korea. hee.ro.park@samsung.com
Telephone: +82-2-34102612 Fax: +82-2-34102619
Received: March 28, 2018
Peer-review started: March 28, 2018
First decision: May 9, 2018
Revised: May 28, 2018
Accepted: June 25, 2018
Article in press: June 25, 2018
Published online: July 28, 2018

Abstract

Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related death, as few patients can be treated with currently available curative local modalities. In patients with HCC where curative modalities are not feasible, radiation therapy (RT) has emerged as an alternative or combination therapy. With the development of various technologies, RT has been increasingly used for the management of HCC. Among these advances, proton beam therapy (PBT) has several unique physical properties that give it a finite range in a distal direction, and thus no exit dose along the beam path. Therefore, PBT has dosimetric advantages compared with X-ray therapy for the treatment of HCC. Indeed, various reports in the literature have described the favorable clinical outcomes and improved safety of PBT for HCC patients compared with X-ray therapy. However, there are some technical issues regarding the use of PBT in HCC, including uncertainty of organ motion and inaccuracy during calculation of tissue density and beam range, all of which may reduce the robustness of a PBT treatment plan. In this review, we discuss the physical properties, current clinical data, technical issues, and future perspectives on PBT for the treatment of HCC.

Key Words: Radiation therapy, Hepatocellular carcinoma, Proton beam therapy, X-ray therapy

Core tip: Radiation therapy (RT) is being increasingly utilized for the treatment of hepatocellular carcinoma (HCC). As RT technology develops, proton beam therapy (PBT) has emerged as a method that affords dosimetric advantages compared with X-ray therapy due to its physical properties, including lack of an exit dose along the beam path. Clinical experience with PBT for HCC is accumulating rapidly, and the effectiveness and safety of PBT has been validated. In this review, we discuss the physical properties, current clinical data, technical issues, and future perspectives on PBT for the treatment of HCC.



INTRODUCTION

Hepatocellular carcinoma (HCC) is the most common primary cancer of the liver, accounting for 90% of all liver cancers[1]. HCC is the fifth most common cancer worldwide and the second leading cause of cancer-related death[2]. Although there is a relatively high incidence of HCC in South-East Asia, rates have also been increasing in North America and Western Europe[3]. HCC has various etiological factors including hepatitis B and C infection, alcohol consumption, liver cirrhosis, non-alcoholic fatty liver disease, obesity, diabetes, aflatoxin exposure, and hereditary disorders such as hemochromatosis and alpha-1-antitrypsin deficiency[4-6].

Although there have been improvements in the management of HCC[7,8], application of curative local modalities such as surgical resection, transplantation, and radiofrequency ablation are often limited due to tumor extent, tumor location, or other patient-related factors[9-11]. Therefore, the prognosis of HCC continues to be unsatisfactory, with a five-year survival of less than 20%, despite improvement in overall survival (OS) by non-curative modalities including chemoembolization and sorafenib[12]. For this reason, radiation therapy (RT) as a local modality has emerged as an alternative/combination treatment approach[13-15]. As the prognosis of HCC depends on sustained control of tumor size[16], there has been growing interest in use of external beam RT[13,14,17,18]. More specifically, advances in proton beam therapy (PBT) have brought additional innovation to the field of RT for the treatment of HCC[12,19,20].

PBT has a dosimetric advantage compared to X-ray therapy due to the physical properties of the technique[21,22]. Therefore, PBT is potentially more beneficial in sparing organs-at risk (OARs), especially for cases where the tumor is proximal to OARs. For liver tumors, the tolerance of surrounding normal liver, biliary tracts, and gastrointestinal (GI) structures is the main limiting factor for dose escalation[23,24]. For these reasons, there has been increasing interest in the use of PBT for treating liver tumors, and clinical experiences and evidences regarding the advantages of PBT continue to accumulate[12,20,25,26]. Here, we review the current knowledge and future perspectives on PBT for HCC.

PHYSICAL CHARACTERISTICS AND DOSIMETRIC ADVANTAGES OF PBT

The deposition of X-ray dose decreases gradually along the beam path with increasing beam depth[27]. As a result, an exit dose is inevitably deposited in adjacent normal tissues. Intensity-modulated RT (IMRT) or volumetric-modulated arc therapy (VMAT) techniques can be used to obtain more conformal dose distributions; however, these methods are still unable to avoid low-dose deposition at the distal area of the beam path. Because of these unfavorable characteristics, RT has had a very limited role in the treatment of HCC in patients whose livers are mostly cirrhotic or poorly functioning, as these patients are most vulnerable to radiation-induced liver disease (RILD)[28,29].

Compared to X-ray beams, a proton beam has a finite range of energy deposition and loses most of its energy within a very short distance at the end of the beam range (Figure 1). These results in a sharp rise and fall in energy absorption, known as the Bragg peak. Bragg peaks can be superposed to provide wider depth coverage, so-called “spread-out Bragg peak beams (SOBP)”[27]. Furthermore, due to the absence of an exit dose, PBT has dosimetric advantages compared with 3-dimentional conformal RT or even IMRT or VMAT[21,22]. Therefore, PBT has the potential to both decrease the risk of RILD and allow for safer escalation of radiation dose[30].

Figure 1
Figure 1 Radiation dose distribution according to technique of radiation therapy. Axial views of A: 3-dimentional conformal radiation therapy (3D-CRT); B: Volumetric arc therapy (VMAT); C: Passive scattering proton beam therapy (PBT); D: Pencil beam scanning PBT; sagittal views of E: 3D-CRT; F: VMAT; G: PBT with wobbling technique; H: PBT with pencil beam scanning technique. There are low dose distributions in 3D-CRT (A, E) and VMAT (B, F) due to the exit dose.
BIOLOGICAL CHARACTERISTICS OF PBT

The proton beams cause deoxyribose-nucleotide acid (DNA) damage and cytotoxicity by direct collisions with DNA molecules and by the generation of radical oxygen specimen (ROS) causing indirect DNA damage[31]. The efficiency of DNA damage is better for PBT than X-ray therapy due to relatively high energies of PBT used for RT[32]. Therefore, the PBT is more effect for a given prescription dose than X-rays[33]. The concept of the relative biological effectiveness (RBE) is useful to consider this issue. The definition of RBE is the ratio of the proton dose to the photon dose for a given level of effect, and proton has higher value of RBE than X-ray. The RBE of the proton beams depends on various parameters such as dose, considered endpoint, linear energy transfer, α/β of the tumor or tissue, and the positions of the SOBP[34]. There is trends that the RBE increases as decreasing α/β, decreasing dose, increasing LET, and approaching the distal edge and distal fall-off of the SOBP[35-37]. Because these dependencies are considered negligible compared with the uncertainties for tissue density[33], beam ranges, and patients, up to date, fixed value of 1.1 which is based on various in vivo and in vitro data has been commonly used as universal value of RBE for proton beam[38]. However, the clinical impact of variability in RBE has been being investigated and with precise modeling variable RBE can be used for further optimization of PBT plan[33].

CLINICAL OUTCOMES OF PBT FOR HCC

The majority of clinical outcomes regarding PBT for HCC have been reported from Eastern Asia and the United States (US) due to the high incidence of HCC and long-term experience in the clinical use of PBT [19,20]. An overview of the published studies of PBT for HCC is summarized in Table 1. The Proton Medical Research Center (PMRC) of the University of Tsukuba, Japan, developed three PBT protocols according to tumor location with respect to the porta hepatis and GI OARs[30,39-42]. Specifically, for peripheral tumors > 2 cm from both GI tract and the porta hepatis, a 66-Gy relative biologic equivalent (GyE) in 10 fractions is delivered. Likewise, tumors within 2 cm of the GI tract can be treated with 77.0 GyE in 35 fractions, while those within 2 cm of the porta hepatis are treated with 72.6 GyE in 22 fractions. In those studies, the authors reported local control (LC) in the range of 88% to 95%, which was similar among various dose and fractionations. In addition, the authors reported 3-year OS rates ranging from 45% to 65%[30,39-42]. They also found that liver function was a significant prognostic factor associated with OS rates, and that OS was better in patients with a Child-Pugh (CP) A score compared with CP B.

Table 1 Studies of proton beam therapy for hepatocellular carcinoma.
StudyPatients (n)CTPscoreTumorsize (cm)PVTTRT regimen (GyE/fractions)LCPFSOS
Tsukuba (2008)[39]53A: 87% B: 11% C: 2%≤ 3: 25% > 3 - < 5: 34% ≥ 5 - < 10: 34% ≥ 10: 8%28%72.6/22 (80.5 Gy EQD2)94% at 2 yr 86% at 3 yr38% at 2 yr 25% at 3 yr2-yr 57% 3-yr 45%
Tsukuba (2009)[40]51A: 80% B: 20%Median 2.8 (range, 0.8 to 9.3) ≤ 5: 88% > 5: 12%NA66.0/10 (91.3 Gy EQD2)95% at 3 yr 88% at 5 yrNA3-yr 49% 5-yr 39%
Tsukuba (2009)[41]318A: 74% B: 24% C: 2%NA14%66.0/10 (91.3 Gy EQD2): 32.7% 72.6/22 (80.5 Gy EQD2): 26.7% 77.0/35 (78.3 Gy EQD2): 20.8%NANA3-yr 65% 5-yr 45%
Tsukuba (2011)[42]47A: 75% B: 19% C: 6%NA15%72.6/22 (80.5 Gy EQD2): 34.0% 77.0/35 (78.3 Gy EQD2): 27.7%88% at 3 yr 88% at 4 yrIHRFS 1-yr 66% 3-yr 40% 4-yr 17%3-yr 50% 4-yr 34%
Loma Linda (2011)[43]76A: 29% B: 47% C: 24%Mean 5.5 ≤ 2: 7% > 2 - < 5: 45% ≥ 5 - < 10: 43% ≥ 10: 5%5%63.0/15 (74.6 Gy EQD2)70% at 3 yr80%Median 36 mo
NCC (2015)[45]27A: 89% B: 11%≤ 5: 81% > 5: 19%NA60.0/20 (65.0 Gy EQD2): 29.6% 66.0/22 (71.5 Gy EQD2): 25.9% 72.0/24 (78.0 Gy EQD2): 44.4%80% at 3 yr 64% at 5 yr3-yr 17% 5-yr 03-yr 56% 5-yr 42%
Multiple United States institutions (2016)[25]44A: 73% B: 20% NA: 7%Median 5.0 (range, 1.9 to 12.0)30%Median 58.05/15 (67.1 Gy EQD2) (range, 15.1-67.5/15)95% at 2 yr1-yr 56% 2-yr 40%1-yr 77% 2-yr 63%

In the US, researchers at Loma Linda University performed a phase II trial of PBT in 76 patients with HCC and liver cirrhosis[43]. Patients with extrahepatic metastases, tense ascites, or more than three tumors were excluded. The progression-free survival (PFS) at 3 years for patients meeting the Milan criteria was 60%. Liver transplantation was performed in 18 patients whose eventual 3-year OS rate was 70%; however, the OS rate of patients who did not undergo liver transplantation was 10%. In these patients, multivariate analysis identified the Milan criteria as the only independent factor associated with OS.

Researchers from Loma Linda University also recently reported a preliminary analysis of a randomized trial of TACE versus PBT[44]. In that study, a total of 69 patients who were diagnosed of HCC and met the Milan or San Francisco transplant criteria were enrolled and randomized to either TACE (n = 36) or PBT (n = 33). The dose fractionation regimen of PBT was 70.2 GyE in 15 fractions. An interim analysis showed a trend toward improved 2-year LC (88% vs 45%) and PFS (48% vs 31%) favoring PBT, although the difference between the two methods was not statistically significant. Another phase II trial of PBT for HCC (n = 44) and intrahepatic cholangiocarcinoma (n = 37) performed as a multi-center trial in the US[25] reported the use of dose fractionation regimens of intention of 67.6 GyE in 15 fractions for tumors > 2 cm from the porta hepatis and 58.05 GyE in 15 fractions for those within 2 cm of porta hepatis. In that study, the median dose actually delivered was 58.05 GyE (range, 15.1 to 67.5 GyE) in 15 fractions. The LC and OS rates at 2 years were 95% and 63%, respectively, supporting ongoing phase III trials of RBT in HCC.

In South Korea, the National Cancer Center performed a phase I dose-escalation study for PBT in HCC comprising 60 GyE in 20 fractions, 66 GyE in 22 fractions, or 72 GyE in 24 fractions[45]. The complete response (CR) rates of PBT were 62.5%, 57.1%, and 100% according to increasing dose level, respectively. In addition, the 3-year LC rate was 79.9%, which was significantly higher in patients with CR than in those with non-CR (90% vs 40%, P = 0.003). None of the patients experienced liver toxicities greater than grade 2.

PBT FOR HCC IN SPECIAL SITUATIONS

Although various clinical studies have demonstrated the effectiveness and safety of PBT in the treatment for HCC, the indications for PBT have not yet been determined and continue to evolve. Because of dosimetric advantages, PBT has the potential to be applicable to more complicated cases ineligible for X-ray therapy, such as those with a history of hepatic RT, vascular invasion of the tumor, extremely large tumor burden, and/or poor liver function.

RE-IRRADIATION WITH PBT FOR HCC

RT has played a role as a salvage treatment for recurrent HCC refractory or ineligible for other modalities[46-48]. RT can also be applied to recurrent HCC previously irradiated when there is no appropriate local salvage modality. However, despite the development of modern RT techniques, re-irradiation in HCC remains a challenging issue due to the risk of RILD and GI toxicity[49-51]. Therefore, very few studies have evaluated the feasibility and effectiveness of hepatic re-irradiation for HCC[51]. Nevertheless, PBT has been investigated as a salvage modality for previously irradiated HCC. Scientists at PMRC reported on the outcomes of repeated PBT for HCC[52]. In that study, 27 patients with 68 lesions received 2 or more courses of PBT, and the median interval between the first and second courses was 24.5 mo (range, 3.3 to 79.8 mo). Likewise, the median dose was 72 Gy in 16 fractions and 66 Gy in 16 fractions for the first and subsequent courses, respectively. In the same study, the LC and OS rates were 87.8% and 55.6%, respectively. Based on these data, the authors concluded that for patients with peripheral tumors and CP A liver function, repeated PBT is safe. More recently, researchers at PMRC reported the results of repeated PBT for 83 patients with HCC, specifically including patients who received 3 (n = 12) or 4 (n = 3) courses of PBT[53]. The median doses for the first, second, third, and fourth courses were 71.0, 70.0, 70.0, and 69.3 GyE, respectively, and the 5-year OS rate was 49.4%. Neither severe acute toxicity nor RILD was observed during the study period.

PBT FOR HCC WITH PORTAL VEIN TUMOR THROMBOSIS

Portal vain tumor thrombosis (PVTT) at the time of diagnosis is present in 30%-40% of all patients with advanced HCC[11], and PVTT is a poor prognostic factor with a median survival of 2.7 mo to 4.0 mo[54,55]. Currently, the Barcelona-Clinic Liver Cancer (BCLC) system recommends sorafenib as the only standard treatment in such patients[9]. However, the response rate to sorafenib ranges from 2% to 5%, and the median time to progression is only 2.8 mo[56]. Therefore, efficient local modalities leading to improve clinical outcomes are required. Unfortunately, no local modalities have been proven to be ef–ficient for HCC with PVTT. Surgery and TACE alone are not typically feasible because most cases of HCC with PVTT are unresectable, and the efficacy of TACE is limited due to the aortoportal shunt[57]. However, various studies have reported the outcomes of RT with or without TACE, with an objective response rate ranging from 40% to 60%. Furthermore, among responders, the median PFS is reported to be 15 mo to 20 mo[58-61].

The outcomes of PBT for HCC with PVTT have also been evaluated by PMRC. That study analyzed 12 HCC patients with PVTT in which a median dose regimen of 55 GyE in 10-22 fractions was used (range of 50 to 72 GyE in 10-22 fractions). Radiographic CR was achieved in 6 patients, and there was recanalization of the portal vein in 5 patients. Furthermore, the 2- and 5-year OS rates were 88% and 58%, respectively.

Lastly, the National Cancer Center of Korea reported on the outcomes of PBT using a simultaneous integrated boost technique for HCC with PVTT[62]. The dose regimens were determined depending on the distance of the tumor from GI OARs, and most tumors were treated with 50-66 GyE in 10 fractions while areas closer to an OAR were treated with 30 GyE in 10 fractions. The LC and OS rates reported in that study at 2 years were 88.1% and 51.1%, respectively. In addition, patients treated with equivalent dose of 2 Gy fractions for α/β = 10 Gy (EQD210) of ≥ 80 GyE, tended to show better tumor vascular thrombosis response, LC, and OS.

PBT FOR LARGE HCCs

Tumor size is critical in determining treatment modalities in HCC[63]. Patients with large tumors are not candidates for ablation therapies or liver transplantation[64-66]. Although surgery seems to have role in the treatment of HCCs larger than 10 cm, few of these tumors are actual candidates for resection[67-69]. In such cases, RT represents an alternative modality for large tumors; however, it is not possible to avoid irradiating large volumes of normal liver tissues, which in turn limits sufficient dose escalation[70,71]. Importantly, PBT has the potential to overcome this limitation. Researchers at the University of Hokkaido performed dosimetric comparison of spot-scanning PBT vs IMRT for HCC[72]. For gross tumors of a nominal diameter more than 6.3 cm, the average risk of RILD according to the Lyman-normal-tissue complication probability model was estimated as 94.5% for IMRT compared to 6.2% for PBT. PMRC also reported the clinical result of PBT for 22 patients with HCC of size > 10 cm[73]. In that study, which employed a median dose regimen of 72.6 GyE in 22 fractions (range, 47.3 to 89.1 GyE in 10-35 fractions), the median tumor size was 11 cm (range, 10 cm to 14 cm). The LC and OS at 2 years were 87% and 36%, respectively. RILD occurred in 5 of 22 patients, 3 of whom showed CR.

PBT FOR HCC PATIENTS WITH POOR LIVER FUNCTION

HCC patients with cirrhotic livers or poor liver function have a dismal prognosis with a median survival time of 3 to 9 mo[55]. Although poor liver function itself is a poor prognostic factor, there are limitations in applying local modalities to those patients because it is difficult to preserve the function of the residual untreated liver[74]. Researchers at PMRC reported on the clinical outcomes of PBT for 19 patients with HCC and CP C[75]. The median RT dose was 72 GyE in 16 fractions (range, 50 to 84 GyE in 10 to 24 fractions). The crude LC rate was 95% over a median follow-up of 17 mo, and the OS at 2 years was 42%. In addition, there were no therapy-related toxicities of grade ≥ 3 or deterioration of CP score. Among the total cohort, 14 patients exhibited an improved CP score. Researchers at PMRC also reported on the feasibility of PBT for HCC patients with uncontrollable ascites[76]. Specifically, they used PBT of 24 GyE in a single fraction, and performed precise adjustment of PBT immediately before irradiation. Although their studies only included three patients, objective responses were achieved for all the irradiated tumors, and there were no therapy-related toxicities higher than grade 3.

HEPATIC TOXICITY

RILD is seldom observed in the era of PBT for HCC, and there is very limited literature on PBT-related RILD. Researchers at PMRC evaluated 60 HCC patients treated with PBT consisting of 76 GyE in 20 fractions[77]. Seventy-eight percent of the patients had CP-A liver function, and 82% had liver cirrhosis; prior liver directed therapy had been performed in 60% of the patients. In that study, RILD was defined as anicteric ascites or asterixis. A total of 11 patients exhibited RILD, and seven patients died due to RILD. In addition, they reported that there were no cases of PBT-related RILD for patients with pretreatment indocyanine green retention at 15 min (ICG R15) less than 20%. On the other hand, RILD-related mortality of patients with a pretreatment ICG R15 over 50% was 75%. For patients with pretreatment ICG R15 of 20%-49%, V30GyE < 25% was identified as a predictor for PBT-related RILD. In more recent studies, changes in CP score were reported. Specifically, PMRC evaluated CP scores before and after PBT for 259 HCC patients[24], and found that among the 76% of patients with CP-A, 73% had cirrhosis of the liver and 63% had previously received liver directed treatment. In addition, the rates in the increase in point 1 CP score compared to baseline were 16% and 8% at 1 and 2 years, respectively. Likewise, the rates of point 2 from baseline CP sore were 11% and 22% at 1 and 2 years, respectively. On the other hand, literature from the US and South Korea indicate that only 4% of study patients have an increased CP score after PBT[25,43,78].

GT TRACT TOXICITY

The stomach, duodenum, and bowel are critical OARs because of their proximity to HCC tumors and the vulnerability of patients to radiation toxicity due to portal hypertension-induced gastroduodenopathy[79,80]. A group of researchers from Loma Linda University reported that the incidence of GI toxicities ≥ grade 2 is 7% following RT[43]. Similarly, researchers at PMRC reported an incidence of ≥ grade 2 GI toxicities of 1%-2% in various populations[39,41,81]. For central tumors abutting the GI tract, however, the incidence of GI toxicities ≥ grade 2 increases to 8.5%, with all of the reported forms of GI toxicity involving hemorrhage[42]. Usually, PBT for HCC proximal to GI tract is prone to GI toxicity. Especially, GI tract located near the distal edge of SOBP is potentially vulnerable to severe toxicity because of the uncertainty of the proton beam range and the high RBE value at the distal edge and the fall off of the SOBP. Although there is no specific report regarding of the relation between the variation of RBE and the GI toxicities, several literatures reported the distal edge effects of PBT on toxicities of brain stem[82,83]. Therefore, special caution is necessary when the location of GI tract is near the distal edge of SOBP. Surgical insertion of tissue expander to displace GI tract away from target is considerable in that situation[84].

BILIARY AND OTHER GI TRACT TOXICITY

The biliary tract is an OAR that must be protected during liver RT. Indeed, several studies have reported symptomatic biliary inflammation with or without stricture in tumors related to the biliary system[85,86]. However, some studies have reported that RT for HCC can be safe with respect to biliary toxicity, even in tumors adjacent to the central biliary system[87,88]. For PBT in HCC, data on biliary complications is limited. Researchers at PMRC reviewed 162 patients who underwent PBT for HCC with a median RT dose of 72 GyE in 16 fractions[81]. Among the total cohort, three patients exhibited signs of biliary toxicity such as stenosis of the common bile duct (n = 1), and biloma with infection adjacent to the irradiated volume (n = 2).

CHEST WALL TOXICITY

As PBT allows for dose escalation in the treatment of HCC, the chest wall is frequently irradiated with increasing high doses. In tumors proximal to the chest wall, there is potential risk of chest wall-related toxicities[19]. Although PBT-related chest wall toxicity data are limited, there have been some published studies. Researchers at PMRC reported that the incidence of rib fracture is 16% after PBT of 66 GyE in 10 fractions in 67 HCC patients[89]. Specifically, the authors found that V60 at a biologically effective dose with α/β = 3 is the most statistically significant parameter for estimating the risk of rib fracture after hypofractionated PBT. In addition, researchers at the MD Anderson Cancer Center reported the chest wall toxicities of 135 patients with lung or hepatobiliary cancer who were treated with ≥ 52.5 GyE in 15 fractions of photon therapy or PBT[90]. Among the total cohort, 49 patients received PBT. During the median follow-up of 9 mo, 20 patients had grade 1 chest wall pain, while one patient had grade 2 chest wall pain. Furthermore, a chest wall V40 ≥ 150 cm3 was identified as an independent predictor for chest wall toxicity. Researchers at the University of Washington and other institutions reported another set of data for chest wall toxicity, comprising 37 patients who underwent PBT with a median dose of 60 GyE (range, 35 to 67.5 GyE) in 15 fractions (range, 13 to 20 fractions)[91]. During a median follow-up of 11 mo, chest wall pain of grade ≥ 2 occurred in 30% of patients, with none of the patients exhibiting radiographic evidence of rib fracture. For a 15-fraction regimen, a V47 > 20 cm3, V50 > 17 cm3, and V58 > 8 cm3 were associated with higher rates of chest wall toxicity. Although there is no consensus regarding dose constraints for chest wall toxicities, efforts should be made to reduce chest wall toxicities based on the reported various dose-volumetric parameters in PBT for HCC.

TECHNICAL ISSUES

Proton beams have unique physical properties including a finite range in the distal direction. This range is determined by the density of material in the beam path, and any perturbation in the densities can change the dosimetry of PBT more significantly than compared to traditional photon therapy[92]. This perturbation can be induced by internal organ motion, daily variation in body shape, uncertainties in machine settings, tissue inhomogeneity, and inaccuracies in dosing, ranges, and density calculations[93]. More specifically, the liver is vulnerable to uncertainties of PBT due to organ motion, the interface of air and soft tissue near the diaphragmatic dome, and variations in body shape due to ascites[76].

Various techniques have been investigated to manage the liver-specific problems associated with PBT. In order to overcome uncertainty due to organ motion, the PBT field should ideally encompass the entire range of motion of the tumor; however, this usually results in additional irradiation of normal tissues. Therefore, respiratory gating methods, abdominal compression, or breath hold techniques can be used to reduce the field such that OARs can be protected from unnecessary irradiation[94,95]. Because of the requirement for patient cooperation, identifying patients who are eligible for these techniques remains an important issue.

Techniques for motion control require fine image guidance. However, tumor and surrounding normal liver tissue are often undistinguishable on typical imaging studies such as orthogonal KV X-ray images or cone beam computed tomography[19]. To set a reference location for setup, radio-opaque fiducial markers are often utilized for more reliable and accurate image guidance[96]. Robust optimization is another method for managing imaging uncertainties. One reported algorithm incorporates the uncertainty directly into PBT optimization[97]. Using this algorithm, it is possible to quantify the quality of the treatment plan under certain geometric uncertainties, as well as those from inaccurate calculations of range and tissue density.

FUTURE PERSPECTIVES

Several studies evaluating the role of PT for HCC are ongoing. These studies are aimed at comparing PBT with radiofrequency ablation, transarterial chemoembolization, or sorafenib, which are the current standard treatments recommended according to BCLC staging. The interim results of a randomized trial comparing PBT with transarterial chemoembolization were reported as discussed above[44]. In addition, various single-arm phase II studies of PBT for HCC in specific clinical situations such as PVTT and inoperable disease are ongoing. The results of these studies will provide many answers to ongoing questions and increase the level of evidence in the field of PBT for HCC.

Technologies related to PBT are also under development. Almost all of the clinical data from PBT for HCC have relied on the passive scattering technique[20]. However, recently constructed PBT centers are now utilizing pencil beam scanning nozzles, which allow for manipulation of the intensity of proton therapy[98-100]. Pencil beam scanning shows better dose optimization and proximal edge conformity compared with passive scattering[101,102]. On the other hand, pencil beam scanning takes more time to deliver the proton beam, involves a broader lateral penumbra, and is more sensitive to organ motion[103-105]. Therefore, robustness optimization and evaluation tools, respiration management such as repainting, and fine image guidance are required for the use of pencil beam scanning in the treatment of HCC[106,107]. Although these issues continue to be investigated and studied, the pencil beam scanning method with simultaneous intensity boost technique is now being tried in HCC[108]. Nevertheless, in order to validate the effectiveness and safety of the pencil beam scanning PBT for HCC, large scale prospective trials will be necessary.

Biological issues are another field which can be more advanced in the future. If the RBE variation can be utilized in the optimization of treatment planning, the therapeutic window would be increased[33]. The variability of RBE is actively being investigated regarding the relevance of not only dose range but various value of α/β’s from different tissues[109,110]. The biology of HCC is diverse according to the various genomic aberrations and the genetic association with radiosensitivity of HCC has been also investigated in in vitro and in vivo[111-113]. Although there is no current data for radiaosensitivity of HCC for PBT specifically, the investigations about the tissue specific radiosensitivity which can be represented with RBE will give the opportunity to provide the optimal strategy of tissue-specific PBT. This is the direction of the approach for PBT to be developed as precision medicine for HCC patients.

CONCLUSION

PBT has significant dosimetric advantages compared with X-ray therapy, which has translated to significant differences in clinical outcomes for the treatment for HCC[20]. The American Society for Radiation Oncology includes HCC in the “group I” indications for PBT, meaning that PBT is recognized as an effective and safe local modality for the treatment of HCC. Various studies comparing with other local modalities are ongoing, and we expect that PBT will become a mainstay of local treatment of HCC in the near future.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Gastroenterology and hepatology

Country of origin: South Korea

Peer-review report classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P- Reviewer: Zhao HT S- Editor: Wang JL L- Editor: A E- Editor: Yin SY

References
1.  Ghouri YA, Mian I, Rowe JH. Review of hepatocellular carcinoma: Epidemiology, etiology, and carcinogenesis. J Carcinog. 2017;16:1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 413]  [Cited by in F6Publishing: 486]  [Article Influence: 69.4]  [Reference Citation Analysis (0)]
2.  Lafaro KJ, Demirjian AN, Pawlik TM. Epidemiology of hepatocellular carcinoma. Surg Oncol Clin N Am. 2015;24:1-17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 194]  [Cited by in F6Publishing: 219]  [Article Influence: 21.9]  [Reference Citation Analysis (0)]
3.  Mittal S, El-Serag HB. Epidemiology of hepatocellular carcinoma: consider the population. J Clin Gastroenterol. 2013;47 Suppl:S2-S6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 770]  [Cited by in F6Publishing: 832]  [Article Influence: 75.6]  [Reference Citation Analysis (0)]
4.  Fattovich G, Stroffolini T, Zagni I, Donato F. Hepatocellular carcinoma in cirrhosis: incidence and risk factors. Gastroenterology. 2004;127:S35-S50.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1691]  [Cited by in F6Publishing: 1665]  [Article Influence: 83.3]  [Reference Citation Analysis (2)]
5.  El-Serag HB. Hepatocellular carcinoma. N Engl J Med. 2011;365:1118-1127.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2881]  [Cited by in F6Publishing: 2983]  [Article Influence: 229.5]  [Reference Citation Analysis (0)]
6.  Zhang DY, Friedman SL. Fibrosis-dependent mechanisms of hepatocarcinogenesis. Hepatology. 2012;56:769-775.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 291]  [Cited by in F6Publishing: 299]  [Article Influence: 24.9]  [Reference Citation Analysis (0)]
7.  Toshikuni N, Takuma Y, Tomokuni J, Yamamoto H. Planning Sonography Using Real-time Virtual Sonography and Contrast-enhanced Sonography for Radiofrequency Ablation of Inconspicuous Hepatocellular Carcinoma Nodules. Hepatogastroenterology. 2015;62:661-666.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Zou Q, Li J, Wu D, Yan Z, Wan X, Wang K, Shi L, Lau WY, Wu M, Shen F. Nomograms for Pre-operative and Post-operative Prediction of Long-Term Survival of Patients Who Underwent Repeat Hepatectomy for Recurrent Hepatocellular Carcinoma. Ann Surg Oncol. 2016;23:2618-2626.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 28]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
9.  Forner A, Llovet JM, Bruix J. Hepatocellular carcinoma. Lancet. 2012;379:1245-1255.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3249]  [Cited by in F6Publishing: 3477]  [Article Influence: 289.8]  [Reference Citation Analysis (3)]
10.  Sinn DH, Gwak GY, Cho J, Paik SW, Yoo BC. Comparison of clinical manifestations and outcomes between hepatitis B virus- and hepatitis C virus-related hepatocellular carcinoma: analysis of a nationwide cohort. PLoS One. 2014;9:e112184.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 39]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
11.  Yu JI, Park HC. Radiotherapy as valid modality for hepatocellular carcinoma with portal vein tumor thrombosis. World J Gastroenterol. 2016;22:6851-6863.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 46]  [Cited by in F6Publishing: 46]  [Article Influence: 5.8]  [Reference Citation Analysis (2)]
12.  Dionisi F, Ben-Josef E. The use of proton therapy in the treatment of gastrointestinal cancers: liver. Cancer J. 2014;20:371-377.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
13.  Kalogeridi MA, Zygogianni A, Kyrgias G, Kouvaris J, Chatziioannou S, Kelekis N, Kouloulias V. Role of radiotherapy in the management of hepatocellular carcinoma: A systematic review. World J Hepatol. 2015;7:101-112.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 106]  [Article Influence: 11.8]  [Reference Citation Analysis (2)]
14.  Klein J, Dawson LA. Hepatocellular carcinoma radiation therapy: review of evidence and future opportunities. Int J Radiat Oncol Biol Phys. 2013;87:22-32.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 126]  [Cited by in F6Publishing: 145]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
15.  Cho JY, Paik YH, Park HC, Yu JI, Sohn W, Gwak GY, Choi MS, Lee JH, Koh KC, Paik SW. The feasibility of combined transcatheter arterial chemoembolization and radiotherapy for advanced hepatocellular carcinoma. Liver Int. 2014;34:795-801.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 41]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
16.  Kudo M, Chung H, Osaki Y. Prognostic staging system for hepatocellular carcinoma (CLIP score): its value and limitations, and a proposal for a new staging system, the Japan Integrated Staging Score (JIS score). J Gastroenterol. 2003;38:207-215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 501]  [Cited by in F6Publishing: 535]  [Article Influence: 25.5]  [Reference Citation Analysis (0)]
17.  Corbin KS, Hellman S, Weichselbaum RR. Extracranial oligometastases: a subset of metastases curable with stereotactic radiotherapy. J Clin Oncol. 2013;31:1384-1390.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 140]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
18.  Kim SW, Oh D, Park HC, Lim DH, Shin SW, Cho SK, Gwak GY, Choi MS, Paik YH, Paik SW. Transcatheter arterial chemoembolization and radiation therapy for treatment-naïve patients with locally advanced hepatocellular carcinoma. Radiat Oncol J. 2014;32:14-22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 25]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
19.  Yeung RH, Chapman TR, Bowen SR, Apisarnthanarax S. Proton beam therapy for hepatocellular carcinoma. Expert Rev Anticancer Ther. 2017;17:911-924.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 14]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
20.  Dionisi F, Widesott L, Lorentini S, Amichetti M. Is there a role for proton therapy in the treatment of hepatocellular carcinoma? A systematic review. Radiother Oncol. 2014;111:1-10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 41]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
21.  Wang X, Krishnan S, Zhang X, Dong L, Briere T, Crane CH, Martel M, Gillin M, Mohan R, Beddar S. Proton radiotherapy for liver tumors: dosimetric advantages over photon plans. Med Dosim. 2008;33:259-267.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 69]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
22.  Gandhi SJ, Liang X, Ding X, Zhu TC, Ben-Josef E, Plastaras JP, Metz JM, Both S, Apisarnthanarax S. Clinical decision tool for optimal delivery of liver stereotactic body radiation therapy: Photons versus protons. Pract Radiat Oncol. 2015;5:209-218.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 45]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
23.  Lasley FD, Mannina EM, Johnson CS, Perkins SM, Althouse S, Maluccio M, Kwo P, Cárdenes H. Treatment variables related to liver toxicity in patients with hepatocellular carcinoma, Child-Pugh class A and B enrolled in a phase 1-2 trial of stereotactic body radiation therapy. Pract Radiat Oncol. 2015;5:e443-e449.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 100]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
24.  Mizumoto M, Okumura T, Hashimoto T, Fukuda K, Oshiro Y, Fukumitsu N, Abei M, Kawaguchi A, Hayashi Y, Ohkawa A. Evaluation of liver function after proton beam therapy for hepatocellular carcinoma. Int J Radiat Oncol Biol Phys. 2012;82:e529-e535.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 49]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
25.  Hong TS, Wo JY, Yeap BY, Ben-Josef E, McDonnell EI, Blaszkowsky LS, Kwak EL, Allen JN, Clark JW, Goyal L. Multi-Institutional Phase II Study of High-Dose Hypofractionated Proton Beam Therapy in Patients With Localized, Unresectable Hepatocellular Carcinoma and Intrahepatic Cholangiocarcinoma. J Clin Oncol. 2016;34:460-468.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 258]  [Cited by in F6Publishing: 302]  [Article Influence: 33.6]  [Reference Citation Analysis (0)]
26.  Fukumitsu N, Okumura T, Takizawa D, Makishima H, Numajiri H, Murofushi K, Ohnishi K, Mizumoto M, Aihara T, Ishikawa H. Proton beam therapy for metastatic liver tumors. Radiother Oncol. 2015;117:322-327.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 22]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
27.  Engelsman M, Schwarz M, Dong L. Physics controversies in proton therapy. Semin Radiat Oncol. 2013;23:88-96.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 109]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
28.  Dawson LA, Normolle D, Balter JM, McGinn CJ, Lawrence TS, Ten Haken RK. Analysis of radiation-induced liver disease using the Lyman NTCP model. Int J Radiat Oncol Biol Phys. 2002;53:810-821.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 576]  [Cited by in F6Publishing: 528]  [Article Influence: 24.0]  [Reference Citation Analysis (0)]
29.  Lawrence TS, Robertson JM, Anscher MS, Jirtle RL, Ensminger WD, Fajardo LF. Hepatic toxicity resulting from cancer treatment. Int J Radiat Oncol Biol Phys. 1995;31:1237-1248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 522]  [Cited by in F6Publishing: 464]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
30.  Mizumoto M, Okumura T, Hashimoto T, Fukuda K, Oshiro Y, Fukumitsu N, Abei M, Kawaguchi A, Hayashi Y, Ookawa A. Proton beam therapy for hepatocellular carcinoma: a comparison of three treatment protocols. Int J Radiat Oncol Biol Phys. 2011;81:1039-1045.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 115]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
31.  Alan Mitteer R, Wang Y, Shah J, Gordon S, Fager M, Butter PP, Jun Kim H, Guardiola-Salmeron C, Carabe-Fernandez A, Fan Y. Proton beam radiation induces DNA damage and cell apoptosis in glioma stem cells through reactive oxygen species. Sci Rep. 2015;5:13961.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 97]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
32.  Durante M, Loeffler JS. Charged particles in radiation oncology. Nat Rev Clin Oncol. 2010;7:37-43.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 463]  [Cited by in F6Publishing: 413]  [Article Influence: 27.5]  [Reference Citation Analysis (0)]
33.  Paganetti H. Relating proton treatments to photon treatments via the relative biological effectiveness-should we revise current clinical practice? Int J Radiat Oncol Biol Phys. 2015;91:892-894.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 49]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
34.  Matsumoto Y, Matsuura T, Wada M, Egashira Y, Nishio T, Furusawa Y. Enhanced radiobiological effects at the distal end of a clinical proton beam: in vitro study. J Radiat Res. 2014;55:816-822.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 47]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
35.  Paganetti H. Relative biological effectiveness (RBE) values for proton beam therapy. Variations as a function of biological endpoint, dose, and linear energy transfer. Phys Med Biol. 2014;59:R419-R472.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 539]  [Cited by in F6Publishing: 586]  [Article Influence: 58.6]  [Reference Citation Analysis (0)]
36.  Wouters BG, Lam GK, Oelfke U, Gardey K, Durand RE, Skarsgard LD. Measurements of relative biological effectiveness of the 70 MeV proton beam at TRIUMF using Chinese hamster V79 cells and the high-precision cell sorter assay. Radiat Res. 1996;146:159-170.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 98]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
37.  Carabe A, Moteabbed M, Depauw N, Schuemann J, Paganetti H. Range uncertainty in proton therapy due to variable biological effectiveness. Phys Med Biol. 2012;57:1159-1172.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 160]  [Cited by in F6Publishing: 161]  [Article Influence: 13.4]  [Reference Citation Analysis (0)]
38.  Paganetti H, Niemierko A, Ancukiewicz M, Gerweck LE, Goitein M, Loeffler JS, Suit HD. Relative biological effectiveness (RBE) values for proton beam therapy. Int J Radiat Oncol Biol Phys. 2002;53:407-421.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 652]  [Cited by in F6Publishing: 598]  [Article Influence: 27.2]  [Reference Citation Analysis (0)]
39.  Mizumoto M, Tokuuye K, Sugahara S, Nakayama H, Fukumitsu N, Ohara K, Abei M, Shoda J, Tohno E, Minami M. Proton beam therapy for hepatocellular carcinoma adjacent to the porta hepatis. Int J Radiat Oncol Biol Phys. 2008;71:462-467.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 78]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
40.  Fukumitsu N, Sugahara S, Nakayama H, Fukuda K, Mizumoto M, Abei M, Shoda J, Thono E, Tsuboi K, Tokuuye K. A prospective study of hypofractionated proton beam therapy for patients with hepatocellular carcinoma. Int J Radiat Oncol Biol Phys. 2009;74:831-836.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 157]  [Cited by in F6Publishing: 154]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
41.  Nakayama H, Sugahara S, Tokita M, Fukuda K, Mizumoto M, Abei M, Shoda J, Sakurai H, Tsuboi K, Tokuuye K. Proton beam therapy for hepatocellular carcinoma: the University of Tsukuba experience. Cancer. 2009;115:5499-5506.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 104]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
42.  Nakayama H, Sugahara S, Fukuda K, Abei M, Shoda J, Sakurai H, Tsuboi K, Matsuzaki Y, Tokuuye K. Proton beam therapy for hepatocellular carcinoma located adjacent to the alimentary tract. Int J Radiat Oncol Biol Phys. 2011;80:992-995.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 41]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
43.  Bush DA, Kayali Z, Grove R, Slater JD. The safety and efficacy of high-dose proton beam radiotherapy for hepatocellular carcinoma: a phase 2 prospective trial. Cancer. 2011;117:3053-3059.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 143]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
44.  Bush DA, Smith JC, Slater JD, Volk ML, Reeves ME, Cheng J, Grove R, de Vera ME. Randomized Clinical Trial Comparing Proton Beam Radiation Therapy with Transarterial Chemoembolization for Hepatocellular Carcinoma: Results of an Interim Analysis. Int J Radiat Oncol Biol Phys. 2016;95:477-482.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 93]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
45.  Kim TH, Park JW, Kim YJ, Kim BH, Woo SM, Moon SH, Kim SS, Koh YH, Lee WJ, Park SJ. Phase I dose-escalation study of proton beam therapy for inoperable hepatocellular carcinoma. Cancer Res Treat. 2015;47:34-45.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 50]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
46.  Bae SH, Park HC, Lim DH, Lee JA, Gwak GY, Choi MS, Lee JH, Koh KC, Paik SW, Yoo BC. Salvage treatment with hypofractionated radiotherapy in patients with recurrent small hepatocellular carcinoma. Int J Radiat Oncol Biol Phys. 2012;82:e603-e607.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 27]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
47.  Park JH, Yoon SM, Lim YS, Kim SY, Shim JH, Kim KM, Lee HC, Cho B, Park G, Kim JH. Two-week schedule of hypofractionated radiotherapy as a local salvage treatment for small hepatocellular carcinoma. J Gastroenterol Hepatol. 2013;28:1638-1642.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 22]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
48.  Lim DH, Lee H, Park HC, Lee JA, Kim SW, Yoo BC, Koh KC, Lee JH, Choi MS, Gwak GY. The efficacy of high-dose 3-dimensional conformal radiation therapy in patients with small hepatocellular carcinoma not eligible for other local modalities. Am J Clin Oncol. 2013;36:162-166.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
49.  Abusaris H, Hoogeman M, Nuyttens JJ. Re-irradiation: outcome, cumulative dose and toxicity in patients retreated with stereotactic radiotherapy in the abdominal or pelvic region. Technol Cancer Res Treat. 2012;11:591-597.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 58]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
50.  Lee DS, Woo JY, Kim JW, Seong J. Re-Irradiation of Hepatocellular Carcinoma: Clinical Applicability of Deformable Image Registration. Yonsei Med J. 2016;57:41-49.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 13]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
51.  Seol SW, Yu JI, Park HC, Lim DH, Oh D, Noh JM, Cho WK, Paik SW. Treatment outcome of hepatic re-irradiation in patients with hepatocellular carcinoma. Radiat Oncol J. 2015;33:276-283.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 12]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
52.  Hashimoto T, Tokuuye K, Fukumitsu N, Igaki H, Hata M, Kagei K, Sugahara S, Ohara K, Matsuzaki Y, Akine Y. Repeated proton beam therapy for hepatocellular carcinoma. Int J Radiat Oncol Biol Phys. 2006;65:196-202.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 76]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
53.  Oshiro Y, Mizumoto M, Okumura T, Fukuda K, Fukumitsu N, Abei M, Ishikawa H, Takizawa D, Sakurai H. Analysis of repeated proton beam therapy for patients with hepatocellular carcinoma. Radiother Oncol. 2017;123:240-245.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 29]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
54.  Schöniger-Hekele M, Müller C, Kutilek M, Oesterreicher C, Ferenci P, Gangl A. Hepatocellular carcinoma in Central Europe: prognostic features and survival. Gut. 2001;48:103-109.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 156]  [Cited by in F6Publishing: 167]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
55.  Llovet JM, Bustamante J, Castells A, Vilana R, Ayuso Mdel C, Sala M, Brú C, Rodés J, Bruix J. Natural history of untreated nonsurgical hepatocellular carcinoma: rationale for the design and evaluation of therapeutic trials. Hepatology. 1999;29:62-67.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 839]  [Cited by in F6Publishing: 873]  [Article Influence: 34.9]  [Reference Citation Analysis (1)]
56.  Bruix J, Raoul JL, Sherman M, Mazzaferro V, Bolondi L, Craxi A, Galle PR, Santoro A, Beaugrand M, Sangiovanni A. Efficacy and safety of sorafenib in patients with advanced hepatocellular carcinoma: subanalyses of a phase III trial. J Hepatol. 2012;57:821-829.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 565]  [Cited by in F6Publishing: 615]  [Article Influence: 51.3]  [Reference Citation Analysis (0)]
57.  Lee HS, Kim JS, Choi IJ, Chung JW, Park JH, Kim CY. The safety and efficacy of transcatheter arterial chemoembolization in the treatment of patients with hepatocellular carcinoma and main portal vein obstruction. A prospective controlled study. Cancer. 1997;79:2087-2094.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 4]  [Reference Citation Analysis (0)]
58.  Kim DY, Park W, Lim DH, Lee JH, Yoo BC, Paik SW, Kho KC, Kim TH, Ahn YC, Huh SJ. Three-dimensional conformal radiotherapy for portal vein thrombosis of hepatocellular carcinoma. Cancer. 2005;103:2419-2426.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 151]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
59.  Yu JI, Park HC, Lim DH, Park W, Yoo BC, Paik SW, Koh KC, Lee JH. Prognostic index for portal vein tumor thrombosis in patients with hepatocellular carcinoma treated with radiation therapy. J Korean Med Sci. 2011;26:1014-1022.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 51]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
60.  Yu JI, Yoon SM, Park HC, Kim JH, Kim TH, Park JW, Seong J, Lee IJ, Jang HS, Kay CS. Multicenter validation study of a prognostic index for portal vein tumor thrombosis in hepatocellular carcinoma. Cancer Res Treat. 2014;46:348-357.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
61.  Yoon SM, Lim YS, Won HJ, Kim JH, Kim KM, Lee HC, Chung YH, Lee YS, Lee SG, Park JH. Radiotherapy plus transarterial chemoembolization for hepatocellular carcinoma invading the portal vein: long-term patient outcomes. Int J Radiat Oncol Biol Phys. 2012;82:2004-2011.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 156]  [Cited by in F6Publishing: 172]  [Article Influence: 13.2]  [Reference Citation Analysis (0)]
62.  Kim DY, Park JW, Kim TH, Kim BH, Moon SH, Kim SS, Woo SM, Koh YH, Lee WJ, Kim CM. Risk-adapted simultaneous integrated boost-proton beam therapy (SIB-PBT) for advanced hepatocellular carcinoma with tumour vascular thrombosis. Radiother Oncol. 2017;122:122-129.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 32]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
63.  Bruix J, Sherman M; Practice Guidelines Committee, American Association for the Study of Liver Diseases. Management of hepatocellular carcinoma. Hepatology. 2005;42:1208-1236.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4333]  [Cited by in F6Publishing: 4404]  [Article Influence: 231.8]  [Reference Citation Analysis (0)]
64.  Vilana R, Bruix J, Bru C, Ayuso C, Solé M, Rodés J. Tumor size determines the efficacy of percutaneous ethanol injection for the treatment of small hepatocellular carcinoma. Hepatology. 1992;16:353-357.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 194]  [Cited by in F6Publishing: 198]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
65.  Livraghi T, Goldberg SN, Lazzaroni S, Meloni F, Ierace T, Solbiati L, Gazelle GS. Hepatocellular carcinoma: radio-frequency ablation of medium and large lesions. Radiology. 2000;214:761-768.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 781]  [Cited by in F6Publishing: 817]  [Article Influence: 34.0]  [Reference Citation Analysis (0)]
66.  Haug CE, Jenkins RL, Rohrer RJ, Auchincloss H, Delmonico FL, Freeman RB, Lewis WD, Cosimi AB. Liver transplantation for primary hepatic cancer. Transplantation. 1992;53:376-382.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 97]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
67.  Noguchi T, Kawarada Y, Kitagawa M, Ito F, Sakurai H, Machishi H, Yamagiwa K, Yokoi H, Mizumoto R. Clinicopathologic factors influencing the long-term prognosis following hepatic resection for large hepatocellular carcinoma more than 10 cm in diameter. Semin Oncol. 1997;24:S6-7-S6-13.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Poon RT, Fan ST, Wong J. Selection criteria for hepatic resection in patients with large hepatocellular carcinoma larger than 10 cm in diameter. J Am Coll Surg. 2002;194:592-602.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 127]  [Cited by in F6Publishing: 133]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
69.  Pandey D, Lee KH, Wai CT, Wagholikar G, Tan KC. Long term outcome and prognostic factors for large hepatocellular carcinoma (10 cm or more) after surgical resection. Ann Surg Oncol. 2007;14:2817-2823.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 97]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
70.  McGinn CJ, Ten Haken RK, Ensminger WD, Walker S, Wang S, Lawrence TS. Treatment of intrahepatic cancers with radiation doses based on a normal tissue complication probability model. J Clin Oncol. 1998;16:2246-2252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 121]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
71.  Hawkins MA, Dawson LA. Radiation therapy for hepatocellular carcinoma: from palliation to cure. Cancer. 2006;106:1653-1663.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 174]  [Cited by in F6Publishing: 192]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
72.  Toramatsu C, Katoh N, Shimizu S, Nihongi H, Matsuura T, Takao S, Miyamoto N, Suzuki R, Sutherland K, Kinoshita R. What is the appropriate size criterion for proton radiotherapy for hepatocellular carcinoma? A dosimetric comparison of spot-scanning proton therapy versus intensity-modulated radiation therapy. Radiat Oncol. 2013;8:48.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 48]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
73.  Sugahara S, Oshiro Y, Nakayama H, Fukuda K, Mizumoto M, Abei M, Shoda J, Matsuzaki Y, Thono E, Tokita M. Proton beam therapy for large hepatocellular carcinoma. Int J Radiat Oncol Biol Phys. 2010;76:460-466.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 99]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
74.  Ohara K, Okumura T, Tsuji H, Chiba T, Min M, Tatsuzaki H, Tsujii H, Akine Y, Itai Y. Radiation tolerance of cirrhotic livers in relation to the preserved functional capacity: analysis of patients with hepatocellular carcinoma treated by focused proton beam radiotherapy. Int J Radiat Oncol Biol Phys. 1997;38:367-372.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 35]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
75.  Hata M, Tokuuye K, Sugahara S, Fukumitsu N, Hashimoto T, Ohnishi K, Nemoto K, Ohara K, Matsuzaki Y, Akine Y. Proton beam therapy for hepatocellular carcinoma patients with severe cirrhosis. Strahlenther Onkol. 2006;182:713-720.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 39]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
76.  Hata M, Tokuuye K, Sugahara S, Tohno E, Fukumitsu N, Hashimoto T, Ohnishi K, Nemoto K, Ohara K, Sakae T. Proton irradiation in a single fraction for hepatocellular carcinoma patients with uncontrollable ascites. Technical considerations and results. Strahlenther Onkol. 2007;183:411-416.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 16]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
77.  Kawashima M, Kohno R, Nakachi K, Nishio T, Mitsunaga S, Ikeda M, Konishi M, Takahashi S, Gotohda N, Arahira S. Dose-volume histogram analysis of the safety of proton beam therapy for unresectable hepatocellular carcinoma. Int J Radiat Oncol Biol Phys. 2011;79:1479-1486.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 28]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
78.  Kim JW, Seong J, Lee IJ, Woo JY, Han KH. Phase I dose escalation study of helical intensity-modulated radiotherapy-based stereotactic body radiotherapy for hepatocellular carcinoma. Oncotarget. 2016;7:40756-40766.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 38]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
79.  Chen LS, Lin HC, Hwang SJ, Lee FY, Hou MC, Lee SD. Prevalence of gastric ulcer in cirrhotic patients and its relation to portal hypertension. J Gastroenterol Hepatol. 1996;11:59-64.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 56]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
80.  Kitano S, Dolgor B. Does portal hypertension contribute to the pathogenesis of gastric ulcer associated with liver cirrhosis? J Gastroenterol. 2000;35:79-86.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 47]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
81.  Chiba T, Tokuuye K, Matsuzaki Y, Sugahara S, Chuganji Y, Kagei K, Shoda J, Hata M, Abei M, Igaki H. Proton beam therapy for hepatocellular carcinoma: a retrospective review of 162 patients. Clin Cancer Res. 2005;11:3799-3805.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 198]  [Cited by in F6Publishing: 162]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
82.  Indelicato DJ, Flampouri S, Rotondo RL, Bradley JA, Morris CG, Aldana PR, Sandler E, Mendenhall NP. Incidence and dosimetric parameters of pediatric brainstem toxicity following proton therapy. Acta Oncol. 2014;53:1298-1304.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 142]  [Cited by in F6Publishing: 139]  [Article Influence: 13.9]  [Reference Citation Analysis (0)]
83.  Giantsoudi D, Sethi RV, Yeap BY, Eaton BR, Ebb DH, Caruso PA, Rapalino O, Chen YL, Adams JA, Yock TI. Incidence of CNS Injury for a Cohort of 111 Patients Treated With Proton Therapy for Medulloblastoma: LET and RBE Associations for Areas of Injury. Int J Radiat Oncol Biol Phys. 2016;95:287-296.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 89]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
84.  Komatsu S, Hori Y, Fukumoto T, Murakami M, Hishikawa Y, Ku Y. Surgical spacer placement and proton radiotherapy for unresectable hepatocellular carcinoma. World J Gastroenterol. 2010;16:1800-1803.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 28]  [Cited by in F6Publishing: 32]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
85.  Cherqui D, Palazzo L, Piedbois P, Charlotte F, Duvoux C, Duron JJ, Fagniez PL, Valla D. Common bile duct stricture as a late complication of upper abdominal radiotherapy. J Hepatol. 1994;20:693-697.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 48]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
86.  Halevy A, Adam A, Stamp G, Benjamin IS, Blumgart LH. Radiation stricture of the biliary ducts: an emerging new entity? HPB Surg. 1992;5:267-270.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
87.  Eriguchi T, Takeda A, Sanuki N, Oku Y, Aoki Y, Shigematsu N, Kunieda E. Acceptable toxicity after stereotactic body radiation therapy for liver tumors adjacent to the central biliary system. Int J Radiat Oncol Biol Phys. 2013;85:1006-1011.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 63]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
88.  Yu JI, Park HC, Lim DH, Paik SW. Do Biliary Complications after Hypofractionated Radiation Therapy in Hepatocellular Carcinoma Matter? Cancer Res Treat. 2016;48:574-582.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 13]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
89.  Kanemoto A, Mizumoto M, Okumura T, Takahashi H, Hashimoto T, Oshiro Y, Fukumitsu N, Moritake T, Tsuboi K, Sakae T. Dose-volume histogram analysis for risk factors of radiation-induced rib fracture after hypofractionated proton beam therapy for hepatocellular carcinoma. Acta Oncol. 2013;52:538-544.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 25]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
90.  Swanick CW, Allen PK, Tao R, Olanrewaju AM, Sutton JR, Lin SH, Welsh J, Das P, Chang JY, Crane CH. Incidence and predictors of chest wall toxicity after high-dose radiation therapy in 15 fractions. Pract Radiat Oncol. 2017;7:63-71.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
91.  Yeung R, Bowen SR, Chapman TR, MacLennan GT, Apisarnthanarax S. Chest wall toxicity after hypofractionated proton beam therapy for liver malignancies. Pract Radiat Oncol. 2018;8:287-293.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
92.  Fukumitsu N, Hashimoto T, Okumura T, Mizumoto M, Tohno E, Fukuda K, Abei M, Sakae T, Sakurai H. Investigation of the geometric accuracy of proton beam irradiation in the liver. Int J Radiat Oncol Biol Phys. 2012;82:826-833.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 15]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
93.  Zhang Y, Huth I, Weber DC, Lomax AJ. A statistical comparison of motion mitigation performances and robustness of various pencil beam scanned proton systems for liver tumour treatments. Radiother Oncol. 2018;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 41]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
94.  Hong TS, DeLaney TF, Mamon HJ, Willett CG, Yeap BY, Niemierko A, Wolfgang JA, Lu HM, Adams J, Weyman EA. A prospective feasibility study of respiratory-gated proton beam therapy for liver tumors. Pract Radiat Oncol. 2014;4:316-322.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 38]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
95.  Yu JI, Yoo GS, Cho S, Jung SH, Han Y, Park S, Lee B, Kang W, Sinn DH, Paik YH. Initial clinical outcomes of proton beam radiotherapy for hepatocellular carcinoma. Radiat Oncol J. 2018;36:25-34.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 15]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
96.  Habermehl D, Henkner K, Ecker S, Jäkel O, Debus J, Combs SE. Evaluation of different fiducial markers for image-guided radiotherapy and particle therapy. J Radiat Res. 2013;54 Suppl 1:i61-i68.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 63]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
97.  McGowan SE, Burnet NG, Lomax AJ. Treatment planning optimisation in proton therapy. Br J Radiol. 2013;86:20120288.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 63]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
98.  Lomax AJ, Boehringer T, Coray A, Egger E, Goitein G, Grossmann M, Juelke P, Lin S, Pedroni E, Rohrer B. Intensity modulated proton therapy: a clinical example. Med Phys. 2001;28:317-324.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 198]  [Cited by in F6Publishing: 172]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
99.  Chung K, Han Y, Kim J, Ahn SH, Ju SG, Jung SH, Chung Y, Cho S, Jo K, Shin EH. The first private-hospital based proton therapy center in Korea; status of the Proton Therapy Center at Samsung Medical Center. Radiat Oncol J. 2015;33:337-343.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 38]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
100.  Smith A, Gillin M, Bues M, Zhu XR, Suzuki K, Mohan R, Woo S, Lee A, Komaki R, Cox J. The M. D. Anderson proton therapy system. Med Phys. 2009;36:4068-4083.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 137]  [Cited by in F6Publishing: 125]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
101.  Lomax AJ, Pedroni E, Rutz H, Goitein G. The clinical potential of intensity modulated proton therapy. Z Med Phys. 2004;14:147-152.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 119]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
102.  Giantsoudi D, Grassberger C, Craft D, Niemierko A, Trofimov A, Paganetti H. Linear energy transfer-guided optimization in intensity modulated proton therapy: feasibility study and clinical potential. Int J Radiat Oncol Biol Phys. 2013;87:216-222.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 117]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
103.  Kaderka R, Schardt D, Durante M, Berger T, Ramm U, Licher J, La Tessa C. Out-of-field dose measurements in a water phantom using different radiotherapy modalities. Phys Med Biol. 2012;57:5059-5074.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 61]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
104.  Fava G, Widesott L, Fellin F, Amichetti M, Viesi V, Lomax AJ, Lederer L, Hug EB, Fiorino C, Salvadori G. In-gantry or remote patient positioning? Monte Carlo simulations for proton therapy centers of different sizes. Radiother Oncol. 2012;103:18-24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 16]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
105.  Park PC, Zhu XR, Lee AK, Sahoo N, Melancon AD, Zhang L, Dong L. A beam-specific planning target volume (PTV) design for proton therapy to account for setup and range uncertainties. Int J Radiat Oncol Biol Phys. 2012;82:e329-e336.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 119]  [Cited by in F6Publishing: 117]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
106.  Unkelbach J, Chan TC, Bortfeld T. Accounting for range uncertainties in the optimization of intensity modulated proton therapy. Phys Med Biol. 2007;52:2755-2773.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 173]  [Cited by in F6Publishing: 175]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
107.  Knopf AC, Hong TS, Lomax A. Scanned proton radiotherapy for mobile targets-the effectiveness of re-scanning in the context of different treatment planning approaches and for different motion characteristics. Phys Med Biol. 2011;56:7257-7271.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 141]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
108.  Apisarnthanarax S, Saini J, O’Ryan-Blair A, Castro J, Bowen SR. Intensity Modulated Proton Therapy with Advanced Planning Techniques in a Challenging Hepatocellular Carcinoma Patient. Cureus. 2017;9:e1674.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
109.  Hojo H, Dohmae T, Hotta K, Kohno R, Motegi A, Yagishita A, Makinoshima H, Tsuchihara K, Akimoto T. Difference in the relative biological effectiveness and DNA damage repair processes in response to proton beam therapy according to the positions of the spread out Bragg peak. Radiat Oncol. 2017;12:111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 25]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
110.  Ilicic K, Combs SE, Schmid TE. New insights in the relative radiobiological effectiveness of proton irradiation. Radiat Oncol. 2018;13:6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 39]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
111.  Khemlina G, Ikeda S, Kurzrock R. The biology of Hepatocellular carcinoma: implications for genomic and immune therapies. Mol Cancer. 2017;16:149.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 191]  [Cited by in F6Publishing: 261]  [Article Influence: 37.3]  [Reference Citation Analysis (1)]
112.  Jin Q, Li XJ, Cao PG. miR-26b enhances radiosensitivity of hepatocellular carcinoma cells by targeting EphA2. Iran J Basic Med Sci. 2016;19:851-857.  [PubMed]  [DOI]  [Cited in This Article: ]
113.  Yu L, Sun Y, Li J, Wang Y, Zhu Y, Shi Y, Fan X, Zhou J, Bao Y, Xiao J. Silencing the Girdin gene enhances radio-sensitivity of hepatocellular carcinoma via suppression of glycolytic metabolism. J Exp Clin Cancer Res. 2017;36:110.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]