You are on page 1of 8

Int. J. Radiation Oncology Biol. Phys., Vol. 62, No. 5, pp.

1371–1378, 2005
Copyright © 2005 Elsevier Inc.
Printed in the USA. All rights reserved
0360-3016/05/$–see front matter

doi:10.1016/j.ijrobp.2005.01.002

CLINICAL INVESTIGATION Liver

A PHASE I TRIAL OF STEREOTACTIC BODY RADIATION THERAPY


(SBRT) FOR LIVER METASTASES

TRACEY E. SCHEFTER, M.D.,* BRIAN D. KAVANAGH, M.D., M.P.H.,* ROBERT D. TIMMERMAN, M.D.,†
HIGINIA R. CARDENES, M.D.,‡ ANNA BARON, PH.D.,§ AND LAURIE E. GASPAR, M.D., M.B.A.*
*Department of Radiation Oncology, University of Colorado Health Sciences Center, Aurora, CO; †Department of Radiation
Oncology, University of Texas-Southwestern, Dallas, TX; ‡Department of Radiation Oncology, Indiana University School of
Medicine, Indianapolis, IN; §Department of Biostatics, University of Colorado Comprehensive Cancer Center, Aurora, CO

Purpose: To determine the maximum tolerated dose (MTD) of stereotactic body radiation therapy (SBRT) for
liver metastases.
Methods and Materials: A multicenter Phase I clinical trial was conducted. Eligible patients had one to three liver
metastases, tumor diameter <6 cm, and adequate liver function. The first cohort received 36 Gy to the planning
target volume (PTV) in three fractions (F). Subsequent cohorts received higher doses up to a chosen maximum
of 60 Gy/3F. At least 700 mL of normal liver had to receive a total dose <15 Gy. Dose-limiting toxicity (DLT)
included acute Grade 3 liver or intestinal toxicity or any acute Grade 4 toxicity. The MTD was exceeded if 2/6
patients in a cohort experienced DLT.
Results: Eighteen patients were enrolled (10 male, 8 female): median age, 55 years (range, 26 – 83 years); most
common primary site, colorectal (6 patients); median aggregate gross tumor volume, 18 ml (range, 3–98 ml). Four
patients had multiple tumors. No patient experienced a DLT, and dose was escalated to 60 Gy/3F without
reaching MTD.
Conclusions: Biologically potent doses of SBRT are well tolerated in patients with limited liver metastases.
Results of this study form the basis for an ongoing Phase II SBRT study of 60 Gy over three fractions for liver
metastases. © 2005 Elsevier Inc.

Stereotactic body radiation therapy (SBRT), Liver metastases, Oligometastases, Phase I trial, Hepatic irradiation.

INTRODUCTION noninvasive and also deliverable on an outpatient basis,


Stereotactic body radiation therapy (SBRT) is an emerging with no requirement for anesthesia.
treatment paradigm defined in recently published American The few reported studies of SBRT for liver tumors have
Society for Therapeutic Radiology and Oncology (ASTRO) included both single-fraction and multiple fraction regi-
and American College of Radiology guidelines as a “treat- mens. Herfarth and colleagues at Heidelberg University
ment method to deliver a high dose of radiation to the target, applied single-fraction SBRT to primary and metastatic
utilizing either a single dose or a small number of fractions liver lesions and safely escalated the dose from 14 to 26 Gy
with a high degree of precision within the body” (1). SBRT (8). Blomgren and colleagues at the Karolinska Institute
has previously been labeled with an assortment of monikers administered 20 – 45 Gy in two to four fractions to a group
such as extracranial radiosurgery and extracranial stereotac- of 17 patients with liver metastases, observing only one
tic radioablation. Potential indications for SBRT include a instance of serious toxicity possibly attributable to SBRT
broad spectrum of tumor types and locations (2). (9). Wulf and colleagues at the University of Wurzburg used
Liver metastases provide a logical opportunity for inves- fractionated SBRT, typically 30 Gy in three fractions, in 23
tigating the application of SBRT. Surgical resection of patients with solitary liver lesions and observed no Grade 3
limited metastatic disease in the liver is known to be asso- or higher acute or late toxicity from treatment (10).
ciated with favorable outcome in well-selected patients (3– Our protocol design involves the use of multiple fractions
7), suggesting a clinical benefit from metastectomy in some rather than a single-fraction regimen and is similar to the
patients. SBRT, if demonstrated safe and similarly effec- strategy employed in the Indiana University Phase I trial of
tive, would be advantageous over surgery, being entirely SBRT for medically inoperable lung cancer (11). The ra-

Reprint requests to: Tracey E. Schefter, M.D., Department of Supported (in part) by a grant from the American Cancer Soci-
Radiation Oncology, University of Colorado Health Sciences Cen- ety.
ter, Campus Mailstop F-706, P.O. Box 6510, Denver, CO 80045- Received Dec 1, 2004, and in revised form Jan 2, 2005. Ac-
0510. Tel: (720) 848-0116; Fax: (720) 848-0222; E-mail: cepted for publication Jan 3, 2005.
Tracey.Schefter@uchsc.edu

1371
1372 I. J. Radiation Oncology ● Biology ● Physics Volume 62, Number 5, 2005

tionale for a multiple fraction regimen is based on several volume and was expanded by a minimum 5-mm radial margin and
factors including the well-described putative advantages of 10 mm craniocaudal margin to create the planning target volume
fractionation including reoxygenation of otherwise radiore- (PTV) for patients who were treated using respiratory control. Two
sistant hypoxic tumor and redistribution of tumor clonogens patients were unable to tolerate any type of respiratory control
system, and the PTV was expanded additionally to account for the
into sensitive phase. This Phase I study was designed to
respiratory motion observed at the time of simulation. Extent of
establish the maximum tolerated dose (MTD) of a three- diaphragmatic excursion was evaluated in each patient under flu-
fraction regimen of liver SBRT for patients with limited oroscopy.
liver metastases from solid tumors in anticipation of a Stereotactic body radiation therapy was planned and adminis-
subsequent Phase II study to evaluate the efficacy of SBRT tered using dynamic conformal arcs or multiple noncoplanar static
in that setting. beams generated by a linear accelerator with energies 6 –15 MV.
The nominal prescription dose was prescribed to the isodose line
covering the PTV (generally 80 –90% isodose line). Although
METHODS AND MATERIALS there were no restrictions on the maximum dose delivered within
the PTV itself, normal tissue dose constraints were strictly applied.
Eligibility
Patients were ineligible for enrollment in the SBRT protocol in the
The protocol was approved by the Institutional Review Boards
current dose cohort if the normal tissue dose limits defined below
of the participating institutions—the University of Colorado and
were exceeded.
Indiana University. Patients who had surgically or medically un-
Stereotactic patient repositioning and tumor relocalization was
resectable liver metastases or who declined surgery for resectable
accomplished by referencing fiducial markers on the body immo-
liver metastases were eligible if they met the criteria listed in
bilization device or reflective markers on the patient surface (Ex-
Appendix A.
acTrac, BrainLab, Inc., Westchester, IL). Accurate patient reposi-
Further restrictions were imposed by constraints of the normal
tioning and tumor relocalization were verified with either a repeat
tissue cumulative dose distribution noted in the following section.
verification CT scan of the patient in the treatment position or with
All patients signed a study-specific Institutional Review Board–
the combined application of external fiducial skin markers and
approved consent form before enrollment.
orthogonal X-ray images. Most patients were given prophylactic
antiemetic therapy (generally consisting of a single dose of 5-HT3
Dose escalation inhibitor with or without 4 mg dexamethasone, orally) before daily
Patients received three fractions of SBRT starting at 12 Gy/ SBRT.
fraction (total, 36 Gy), increased by 2 Gy/fraction for each subse- It was considered highly desirable to keep daily treatment times
quent dose group according to a standard Phase I design. Any (total time during which the patient was lying on the treatment
Grade 3 liver, gastric, small bowel, or spinal cord toxicity or any table) to less than 45 min. The rationale was to minimize any
Grade 4 toxicity from SBRT was considered a dose-limiting tox- patient inconvenience or discomfort associated with prolonged
icity (DLT). Each cohort consisted of 3 patients, unless 1 of the immobilization and to avoid the potentially detrimental effects of
patients experienced a DLT, in which case the cohort was ex- intrafraction radiation repair that can occur during excessively
panded to 6 patients. The maximum tolerated dose (MTD) was lengthy individual treatments (12, 13). To comply with this guide-
defined as the dose level below that which results in DLT in 2 or line, some patients with multiple lesions were treated with two
more of the 6 patients in each cohort. An upper limit of 20 separate three-fraction treatment courses. The three fractions of
Gy/fraction (total dose, 60 Gy in three fractions) was preselected SBRT administered to any given lesion could be given on consec-
at the time of study design as the highest dose level to be evalu- utive days or could be separated with intervening days, according
ated. It was determined that at least 5 patients would be enrolled in to the preference of the treating radiation oncologist and practical
this cohort even if none of the first 3 experienced DLT, as a means clinic scheduling considerations; the total course of SBRT was to
of added verification of dose tolerance. be completed within 14 days.

SBRT technique and dose specifications Normal tissue dose constraints


Patients were immobilized during computed tomography (CT) To establish specified limitations on the dose to normal liver, we
simulation and treatment using a Stereotactic Body Frame (Elekta, applied a strategy concordant with the so-called “critical volume
Inc., Crawley, UK) or equivalent customized external vacuum- model” for normal tissue injury originally proposed by Yaes and
type or synthetic body mold. Respiratory control was achieved Kalend (14). Published reports have indicated that up to 80% of the
using an Active Breathing Coordinator (Elekta, Inc.) or abdominal liver can be safely removed in a patient with adequate liver
compression whenever possible during both simulation and treat- function (15). To exercise an even greater level of caution in terms
ment. In some cases, a set of planning CT images through the liver of the volume of normal liver tissue that must be preserved,
only were obtained using rapid acquisition after intravenous ra- estimating that a typical normal liver volume is approximately
diographic contrast material infusion to highlight the tumor; an- 2000 mL, we specified that a minimum volume of 700 mL or 35%
other set of images covering the liver plus a generous margin of normal liver should remain uninjured by SBRT. The tolerance
above and below were obtained using the respiratory control dose when the entire liver is irradiated has been demonstrated to be
device. These two sets of images were then fused, often with at least 33 Gy in 22 fractions (16). For a generic alpha/beta ratio
additional pre-SBRT diagnostic CT images, magnetic resonance estimate of 3 Gy and the assumption of no tumor proliferation
images. or positron emission tomography-CT scan, to allow accu- during treatment, the biologically equivalent dose of this schedule
rate identification of the location of the gross tumor volume (GTV) of 33 Gy in 22 fractions is 49.5 Gy3 according to standard
during treatment. linear-quadratic modeling, ignoring any proliferation correction
The GTV was considered to be identical to the clinical target (17). An SBRT regimen of 15 Gy in three fractions would have a
A Phase I trial of SBRT ● T. E. SCHEFTER et al. 1373

normal tissue biologically equivalent dose of 40 Gy3, safely below were in close proximity, more than one GTV was included
the maximum tolerable level. We therefore mandated that at least within the same PTV. The median aggregate total gross tumor
700 mL of normal liver (entire liver minus cumulative GTV) had volume (sum of all GTVs) was 17.8 mL (range, 3–98 mL).
to receive at total dose less than 15 Gy. PTVs ranged in magnitude from 8 –210 mL (median, 41 mL).
Furthermore, at least 67% of the right kidney had to receive a
The primary sites of disease are listed in Table 1. Two patients
total dose of less than 15 Gy in three fractions (5 Gy per fraction).
The percent of total kidney volume (defined as the sum of the left
with primary hepatocellular carcinoma were treated. The
and right kidney volumes) receiving 15 Gy total in three fractions first was treated in the 36-Gy cohort as definitive therapy for
(5 Gy per fraction) was required to be less than 35% of the total a patient with medically inoperable disease. Subsequently,
kidney volume. The maximum dose to any point within the spinal the investigators elected to write a separate Phase I protocol
cord could not exceed 18 Gy total in three fractions (6 Gy per for patients treated with SBRT as primary therapy for hep-
fraction), and the maximum point dose to the stomach or small atocellular carcinoma. The second hepatocellular carcinoma
intestine could not exceed 30 Gy total in three fractions (10 Gy per patient on the current protocol was treated for a focus of
fraction). hepatic metastasis separate from his previously resected
primary cancer.
Follow-up Sixteen patients had received at least one type of systemic
Patients were evaluated at least once during the treatment course therapy before liver SBRT; 8 patients had received three or
for detailed account of new symptoms. Any observed toxicity at more types of chemotherapeutic agent. The most common
this time and at all other times was documented in the case report agents were a platinum derivative (10 patients), a taxane (6
form. Additional follow-up visits were planned at 4 – 6 weeks after patients), and 5-fluorouracil (4 patients). One patient had
treatment completion and at 3-month intervals for the first 2 years
gefitinib alone before SBRT. The patient with ovarian can-
after treatment completion. Serum liver enzymes were obtained
cer had had prior radiofrequency ablation to a different liver
4 – 6 weeks after SBRT and at 3-month intervals. Physical exam-
ination, including abdominal inspection and palpation, was per- metastasis. Twelve patients remained alive at the time of
formed at each follow-up visit. Documentation at each visit in- analysis, a median of 7.1 months after enrollment in the
cluded performance status, fatigue and general malaise, skin protocol (range, 3.8 –12.3 months). Six patients died within
toxicity, pain, anorexia, nausea, vomiting, dyspepsia, and any 3.1–18.9 months after enrollment. One patient died most
other symptoms reported by the subject. Liver imaging was ob- likely as a result of progressive liver metastases, 4 patients
tained at 3-month intervals. died of complications from progressive extrahepatic metas-
All data were stored and managed using Research Data Man- tases, and 1 patient died from preexisting medical comor-
agement Framework (www.RDMF.net, Aurora, CO). System con- bities unrelated to SBRT.
sulting and data management were provided by Whitedragonfly
Systems, LLC, Aurora, CO.
Toxicity
RESULTS The patient groups, number of patients per dose group,
and all toxicity are listed in Table 2. No patient experienced
Patient population dose-limiting toxicity. A fourth patient was enrolled in the
A total of 18 patients were enrolled in the protocol: 10 fourth group, total dose 54 Gy, because he was unwilling to
men and 8 women. The median age at the time of enroll- wait until the mandated 30-day observation time after treat-
ment was 54 years (range, 26 – 83 years). The median time ment of the third patient in the same group before dose
from initial cancer diagnosis to SBRT study enrollment was escalation. It was reasoned that if the third patient had
19 months (range, 6 –232). Fourteen patients had a solitary experienced a DLT, the group would have expanded, anyway;
lesion, 2 had two lesions, and 2 patients had three individual if the fourth patient had experienced DLT, the group would
discrete lesions. In some instances in which multiple GTVs have been expanded to 6 patients. Because the 60-Gy total
dose level was the highest planned dose escalation, 5 patients
were enrolled. There were no observed cases of DLT; there-
Table 1. Primary site of malignancy for patients treated on the fore, it was concluded that the MTD had not been exceeded.
Phase I SBRT protocol
Prophylactic antiemetic therapy was allowed at the dis-
Primary site n cretion of the treating radiation oncologist. Twelve patients
were premedicated: 8 received granisetron, 1 of whom also
Bladder 1 received dexamethasone; 3 received ondansetron, 1 of
Breast 1
Colorectal 6
whom also received dexamethasone; and 1 patient received
Esophageal 1 dexamethasone alone. No patient complained of nausea
Liver* 2 after liver SBRT. Serial evaluation of liver function tests
Head and neck 1 revealed no significant changes in bilirubin or alkaline phos-
Lung 4 phatase levels (data not shown). Among patients not known
Ovary 1
Pancreas 1
to have disease progression within the liver, there was a
trend toward modest elevation of alanine aminotransferase
* See text for explanation. and aspartate aminotransferase, as shown in Fig. 1.
1374 I. J. Radiation Oncology ● Biology ● Physics Volume 62, Number 5, 2005

Table 2. Patient groups and number of patients per group (n)

Dose per Total dose


Group n fraction (Gy) (Gy) DLTs Non-DLT toxicity

1 3 12 36 None
2 3 14 42 None
3 3 16 48 None Grade 1 dermatitis *(1)
4 4 18 54 None Grade 1 pain (1)
5 5 20 60 None Grade 1 fatigue (1), grade 1
dermatitis* (1), grade 1 pain (1)

There were no dose-limiting toxicities (DLTs) observed in any patient. See text for explanation of group size.
* Dermatitis was limited to localized erythema and hyperpigmentation with minimal dry desquamation, as previously described (18).

Normal tissue dosimetry After SBRT, it was typical to observe radiographic


In 11 patients, the maximum dose per fraction at any changes in the liver of the nature and appearance described
point in the spinal cord was less than 2 Gy and was not by Herfarth and colleagues (19), namely well-circumscribed
further analyzed; in the remaining 7 patients, the maximum hypodense regions recapitulating the dose distribution. Fig-
dose at any point within the spinal cord ranged from 3.3– 6.0 ures 3 and 4 illustrate the isodose distributions from two
Gy per fraction (9.9 –18 Gy total dose). In all of these cases, representative cases and changes observed on follow-up
the volume of spinal cord receiving a cumulative total dose imaging. The post-treatment hypodense region corre-
above 15 Gy was less than 1 mL. sponded roughly to the volume of uninvolved liver that
In 12 patients, the maximum cumulative dose to any received 30 Gy or more in three fractions.
point in the right kidney was less than 15 Gy, ranging from
0.1–12 Gy. In the 6 patients in whom a portion of the right Prescription isodose, target coverage, and equivalent
kidney received a cumulative dose greater than 15 Gy uniform dose analysis
through the three fractions administered, the median percent The isodose distribution applied during SBRT typically
of the right kidney that received more than 15 Gy was 16% includes planned heterogeneity within the tumor intended to
(range, 2–36). Doses administered to the left kidneys were intensify the dose within the tumor (20). In this protocol, the
even lower, as expected, and are not further analyzed since median isodose line prescribed to cover the PTV was 80%
there were no clinical consequences from dose to the kidneys. of the maximum point dose within the PTV (range, 72–
The median volume of uninvolved liver (normal liver 88%). The median percent of the PTV covered by the
minus GTV) was 1783 mL (range, 875–2806 mL). The prescription isodose line was 98% (range, 91–100%).
median volume of uninvolved liver that received a cumu- To evaluate the effect of dose inhomogeneity within the
lative dose of less than 15 Gy in three fractions was 1362 target volumes on the equivalent uniform dose (EUD) ad-
mL (range, 700 –2662 mL). The dose–volume histograms
for uninvolved liver for all patients are shown in Fig. 2.
3000

60 2500 Group 1
Group 2
2000 Group 3
uninvolved liver, cc

45
Group 4
1500
Group 5
Units/Liter

30 1000

500
15
AST
ALT 0
0 15 30 45 60
0 dose, Gy
Baseline 1-1.5 mos 2-3 mos
Fig. 2. Absolute dose (Gy) vs. absolute volume of uninvolved liver
Fig. 1. Changes in Alanine aminotransferase (ALT) and aspartate (mL). Patients in different dose cohorts are represented with the
aminotransferase (AST) after stereotactic body radiation therapy. different line styles shown in the figure. Group 1 patients received
The normal range is 0 – 47 U/L. The positive error bars (standard a nominal dose of 36 Gy in three fractions to the planning target
deviation) are shown for ALT, and negative error bars are shown volume; Group 2, 42 Gy in three fractions; Group 3, 48 Gy in three
for AST. The upward trend at the first follow-up evaluation did not fractions; Group 4, 54 Gy in three fractions; Group 5, 60 Gy in
reach statistical significance. three fractions.
A Phase I trial of SBRT ● T. E. SCHEFTER et al. 1375

ministered, we calculated the EUD according to methods


previously described (21), using the following formula:

EUD ⫽ 2Gy
1n 冋兺
1 N

V i⫽1
Di
Vi共SF2兲 2Gy 册
1n共SF2兲

SF2 is the surviving fraction after 2 Gy (here estimated to be


0.5) and Di is the dose given to the ith subvolume Vi within
the total volume, V. The results are shown in Fig. 5, where
the nominal prescription dose is plotted against the EUD
given to the GTVs and PTVs. It may be seen that an
approximately similar ratio between nominal dose and EUD
was maintained throughout all dose levels within the study.

DISCUSSION
Tolerance of liver SBRT
Results of the present Phase I study indicate that a ste-
reotactic body radiation therapy (SBRT) dose of at least 60
Gy in three fractions may be safely administered to patients
with one to three discrete liver metastases, as long as an
adequate volume of normal liver tissue is spared from the
high-dose region. Doses were escalated according to stan-
dard Phase I design, from 36 Gy to 60 Gy (in three frac-
tions) in increments of 6 Gy per cohort. There was no Grade
3 or 4 toxicity; therefore, the MTD was not reached up to
the predefined upper limit of 60 Gy.
In the liver SBRT experience reported from the Karo-
linska Institute, the single instance of serious toxicity
occurred in a patient with a history of gastritis who
experienced an exacerbation after SBRT that might have Fig. 3. (a) Color wash isodose (total dose) distribution for a
been prompted by the treatment (9). We did not observe representative case. The patient was treated in dose Group 4 (54
Gy in three fractions to the planning target volume). The gross
any Grade 2 or higher bowel toxicity in the present study, tumor volume is outlined in purple and planning target volume in
but it should be emphasized that strict normal tissue dose pink. A single dynamic conformal arc technique was used. Note
limits were applied. It is conceivable that for some le- the very small volume of total liver that received more than 15 Gy
sions on the medial surface of the liver, it might not be in three fractions. (b) Left, 6 weeks post-stereotactic body radia-
possible to restrict the maximum point dose to the stom- tion therapy contrast-enhanced computed tomography scan ob-
tained in the early portal-venous phase. Right, super-positioning of
ach or intestinal tissue to 10 Gy or less. We would urge the 30 Gy isodose volume after fusion of the follow-up image with
extreme caution in this setting, and we would not extend the planning computed tomography image. Note that the hypo-
the observations from the present study to imply an dense region corresponds approximately to the 30 Gy total dose
understanding of the stomach or intestinal MTD with volume.
SBRT, because few patients had lesions that closely
approached the gastrointestinal tract. model’s predictions. The mean dose to the liver was a robust
The critical volume model applied to the normal liver predictor of RILD: no patient who received a mean total
dose constraint in the present study of liver SBRT differs liver dose below 31 Gy suffered RILD, supporting a concept
fundamentally from the Lyman normal tissue complication of liver tissue architecture as a parallel arrangement of
probability model that has been employed by others in dose functional subunits (24).
selection for partial liver irradiation using conventional The lack of any RILD in the present series does not
fractionation (23). Observations from the University of contradict these prior predictions and observations from
Michigan group have demonstrated that the Lyman normal series of conventionally fractionated liver irradiation in any
tissue complication probability model is a good predictor of way. We would attribute the lack of RILD after SBRT with
radiation-induced liver disease. Among a large series of a generally low mean dose to the uninvolved liver, as is
patients treated with chemotherapy and partial liver irradi- apparent in Fig. 2. The mean dose to uninvolved liver
ation, 19 of 203 (9%) suffered Grade 3 or higher radiation- remained acceptably low even in the highest dose group (60
induced liver disease (RILD), consistent with the Lyman Gy in three fractions to the PTV), ranging from 3.3 to 23.9
1376 I. J. Radiation Oncology ● Biology ● Physics Volume 62, Number 5, 2005

Gy (median, 15.3 Gy)—all below the threshold expected to


induce RILD.

Normal tissue effects: radiobiologic and


clinical implications
Stereotactic body radiation therapy induced characteristic
hypodense radiographic changes in regions of uninvolved
liver, as originally described by Herfarth after single-frac-
tion SBRT (19). The radiographic change did not correlate
with any clinical endpoint, and there was no distortion of
normal liver anatomy otherwise seen (i.e., no swelling of the
liver and no displacement of liver vessels or bile ducts).
Nevertheless, it is interesting to consider the effect in terms
of the radiobiologic features, which might give a hint of the
underlying etiology. In their analysis, Herfarth and col-
leagues identified a median threshold dose for the observed
normal tissue reaction of 14 Gy at an interval of 2–3 months
after SBRT. The approximate isoeffective dose in three
fractions from our observations was 30 Gy in the cases
analyzed in the present study.
If we consider, then, the possibility that the phenomenon
results from a radiobiologic process that follows conven-
tional linear-quadratic modeling, we can estimate an ␣/␤
ratio for the effect by considering the biologically equiva-
lent dose equation:

BED ⫽ nd 1 ⫹冉 d
␣⁄␤ 冊
where n ⫽ number of fractions, and d ⫽ dose per fraction.
Setting the isoeffective doses from the Herfarth study and
the present study equal to each other and substituting values

Fig. 4. Planning and follow-up images for another patient treated


in the fourth dose group (54 Gy in three fractions to the planning
target volume). (a) Upper left, three-dimensional rendering of the
liver (pink), kidneys (purple), spinal cord (green), and planning
target volume (brown), with blue and yellow line segments indi-
cating the orientation of the three noncoplanar dynamic conformal
arcs used for treatment. Other panels indicate axial, coronal, and
sagittal views of the gross tumor volume (outlined in green) and
planning target volume (outlined in brown) with surrounding nor-
mal anatomic structures. (b) Upper left, schematic representation
of the start and stop positions of the noncoplanar arcs, here as blue
and yellow fans. Other panels are identical to the corresponding
panels in Fig. 3a, except that the isodose (total dose) distribution is
superimposed as a color wash. (c) Upper panel, pretreatment
portal-venous phase computed tomography showing the large me-
tastasis in the right posterior lobe, with annotated diameter mea-
surement in the slice shown on the left. Center panel, pre–ster-
eotactic body radiation therapy (SBRT) planning image with the
30 Gy isodose contour shown in yellow color wash (left side), with
arrow pointing to the corresponding hypodense region seen at the
3-month post-SBRT follow-up image (right side). Again, the hy-
podense region corresponds to the approximate contours of the 30
Gy isodose volume. Lower panel, 6-month post-SBRT images
showing resolution of the larger hypodense region and minimal
residual abnormality at the site of the treated lesion.
A Phase I trial of SBRT ● T. E. SCHEFTER et al. 1377

80 to have a minimum of 4 – 6 months follow-up interval for


more accurate estimation of in-field tumor control.
70
EUD, GTV SBRT dose delivered to tumor: predicted impact on tumor
EUD, PTV control probability
60
EUD, Gy

The preselected maximum dose level to be tested, 60 Gy


in three fractions, was chosen because it is predicted to
50
provide a very high rate of tumor control. Fowler and
colleagues considered the question of how much dose is
40 necessary to achieve a very high tumor control probability
(TCP) with SBRT, primarily analyzing the use of SBRT for
30 lung cancer, and calculated that a total dose of 60 – 69 Gy in
30 36 42 48 54 60 66 three fractions (20 –23 Gy per fraction) is necessary if there
Nominal prescription dose, Gy is even a small amount of hypoxia present (25). Predictive
Fig. 5. Nominal prescription dose (Gy) vs. equivalent uniform dose models are, of course, only as good as the baseline assump-
(EUD), Gy. Note that a line of identity is also depicted on the tions applied. For example, in the present series the mean
graph. In all except one case, the EUD exceeded the nominal total equivalent uniform dose given to the GTV in the 60
prescription dose. The average ratio of gross tumor volume (GTV) Gy-dose cohort was actually 72 Gy in three fractions, or 24
EUD/prescription dose was 1.2 (range 1.1–1.3). In some cases,
Gy per fraction, and the median GTV was 17 mL. If we
multiple GTVs were encompassed in a single PTV.
apply the same assumptions for estimating TCP that we
have previously employed, namely an estimated clonogen
density of 107/mL and a Gaussian distribution of radiosen-
into the previous equation yields the following expression:
sitivity across the tumor cell population (21), varying the

冉 冊 冉 冊
estimated surviving fraction after 2 Gy from 0.4 to 0.5 will
14 10 alter the predicted TCP for a 17-mL tumor from greater than
14 1 ⫹ ⫽ 30 1 ⫹ 99% down to approximately 75%. It will only be through
␣⁄␤ ␣⁄␤
careful future observations of the actual control rates
achieved with aggressive SBRT that we will be able to
Solving the equation yields an estimate for ␣/␤ of 0.25 Gy. refine our estimates of the true radiosensitivity parameters
Although this very low value would not be expected for in vivo and possibly also understand the influence of tumor
either malignant or normal epithelial tissue, it is not unusual hypoxia in this setting.
for lymphoid cells (22). It is tempting to speculate, then, that
the hypodensity in some way directly or indirectly reflects
local lymphocyte depletion or other local immunomodula- CONCLUSION
tory effect. Our results show that it is safe to deliver 60 Gy in three
Further study would be necessary to establish the mech- fractions of 20 Gy using SBRT techniques for patients with
anistic effect of the observed hypodensity with a higher one to three discrete liver metastases and adequate pre-
level of confidence. However, regardless of the true etiol- SBRT hepatic function, as long as 700 mL of uninvolved
ogy, it is important to be aware of the expected normal normal liver receives less than 15 Gy total dose. This dose
tissue reactions that occur in the few months after SBRT, regimen is predicted to yield a high rate of tumor control. A
because evaluation of local tumor control is more difficult Phase II trial is ongoing to evaluate the rate of local tumor
during that time frame. For fully aggressive liver SBRT to control achieved at an SBRT dose of 60 Gy in three frac-
a dose of 60 Gy in three fractions, it is probably necessary tions.

REFERENCES
1. Potters L, Steinberg M, Rose C, et al. American Society for blood transfusion and determinants of survival after liver
Therapeutic Radiology and Oncology and American College resection for metastatic colorectal carcinoma. Ann Surg
of Radiology practice guidelines for the performance of ste- 1992;216:492–505.
reotactic body radiation therapy. Int J Radiat Oncol Biol Phys 5. Nordlinger B, Parc R, Delva E, et al. Hepatic resection for
2004;60:1026 –1032. colorectal liver metastases. Ann Surg 1987;205:256 –263.
2. Kavanagh BD, Timmerman RD, eds. Stereotactic body radi- 6. Fong Y, Cohen AM, Forter JG, et al. Liver resection for
ation therapy. New York: Lippincott Williams & Wilkins; colorectal metastases. J Clin Oncol 1997;15:938 –946.
2005. 7. Singletary AE, Walsh G, Vauthey JN, et al. A role for curative
3. Gayowksi TJ, Iwatsuki S, Madariaga JR, et al. Experience in surgery in the treatment of selected patients with metastatic
hepatic resection for metastatic colorectal cancer: Analysis of breast cancer. Oncologist 2003;8:241–251.
clinical and pathological risk factors. Surgery 1994;116:703– 8. Herfarth KK, Debus J, Wannenmacher M. Stereotactic radia-
711. tion therapy of liver metastases: Update of the initial phase-I/II
4. Rosen CB, Nagorney DM, Taswell HF, et al. Perioperative trial. Front Radiat Ther Oncol 2004;38:100 –105.
1378 I. J. Radiation Oncology ● Biology ● Physics Volume 62, Number 5, 2005

9. Blomgren J, Lax I, Goranson H, et al. Radiosurgery for tumors 18. Schefter TE, Cardenes HR, Kavanagh BD. Stereotactic body
in the body: Clinical experience using a new method. J Ra- radiation therapy for primary and metastatic liver tumors. In:
diosurg 1998;1:63–74. Kavanagh BD, Timmerman RD, editors. Stereotactic body
10. Wulf J, Hadinger U, Oppitz U, et al. Stereotactic radiotherapy radiation therapy. New York: Lippincott Williams & Wilkins;
of targets in lung and liver. Strahlenther Onkol 2001;177:645– 2005.
655. 19. Herfarth KK, Hof H, Bahner ML, et al. Assessment of focal
11. Timmerman R, Papiez L, McGarry R, et al. Extracranial liver reaction by multiphasic CT after stereotactic single-dose
stereotactic radioablation: Results of a phase I study in med- radiotherapy of liver tumors. Int J Radiat Oncol Biol Phys
ically inoperable stage I non-small cell lung cancer. Chest 2003;57:444 – 451.
2003;124:1946 –1955. 20. Lax I, Blomgren H, Nüslund I, et al. Stereotactic radiotherapy
12. Benedict SH, Lin PS, Zwicker RD, et al. The biological of malignancies in the abdomen: Methodological aspects. Acta
effectiveness of intermittent irradiation as a function of overall Oncol 1994;33:677– 683.
treatment time: Development of correction factors for linac- 21. Kavanagh BD, Timmerman RD, Benedict SH, et al. How
based stereotactic radiotherapy. Int J Radiat Oncol Biol Phys should we describe the radiobiologic effect of extracranial
1997;37:765–769. stereotactic radiosurgery: Equivalent uniform dose or tumor
13. Fowler JF, Welsh JS, Howard SP. Loss of biological effect in control probability? Med Phys 2003;30:321–324.
prolonged fraction delivery. Int J Radiat Oncol Biol Phys 22. Romm H, Stephan G. Dose dependency of FISH-detected
2004;159:242–249. translocations in stable and unstable cells after Cs gamma
14. Yeas RJ, Kalend A. Local stem cell depletion model for irradiation of human lymphocytes in vitro. Cytogenet Genome
radiation myelitis. Int J Radiat Oncol Biol Phys 1988;14: Res 2004;104:162–167.
1247–1259. 23. Dawson LA, Normolle D, Balter JM, et al. Analysis of radi-
15. Penna C, Nordlinger B. Colorectal metastasis (liver and lung). ation-induced liver disease using the Lyman NTCP model. Int
Surg Clin North Am 2002;82:1075–1090. J Radiat Oncol Biol Phys 2002;53:810 – 821.
16. Russell AH, Clyde C, Wasserman TH, et al. Accelerated 24. Jackson A, Ten Haken RK, Robertson JM, et al. Analysis of
hyperfractionated hepatic irradiation in the management of clinical complication data for radiation hepatitis using a par-
patients with liver metastases: Results of the RTOG dose allel architecture model. Int J Radiat Oncol Biol Phys 1995;
escalating protocol. Int J Radiat Oncol Biol Phys 1993;27: 31:883– 891.
117–123. 25. Fowler JF, Tome WA, Welsh JS. Estimation of required doses
17. Fowler JF. The Linear-quadratic formula and progress in in stereotactic body radiation therapy. In: Kavanagh BD, Tim-
fractionated radiotherapy: A review. Br J Radiol 1989;62: merman RD, editor. Stereotactic body radiation therapy. New
679 – 694. York: Lippincott Williams & Wilkins; 2005.

APPENDIX
ELIGIBILITY CRITERIA
● 1–3 liver metastases from any solid tumor except a germ time unless the patient was receiving anticoagulant
cell tumor or lymphoma medication
● Maximum tumor diameter ⬍6 cm ● No chemotherapy within 14 days before or after stereo-
● Karnofsky performance status ⬎60% tactic body radiation therapy
● Life expectancy ⬎3 months ● Age ⱖ18 years
● No prior radiotherapy to the liver ● No evidence of progressive or untreated gross disease
● Adequate liver function, defined as total bilirubin ⬍3 outside of the liver
mg/dL, serum albumin ⬎2.5 g/dL, serum levels of liver ● No active liver infection
enzymes ⬍3 times the upper limit of normal, and ● Adequate kidney function (serum creatinine ⬍1.8 mg/
normal prothrombin time and partial thromboplastin dL)

You might also like