Anda di halaman 1dari 19

G a s t r o i n t e s t i n a l I m a g i n g R ev i ew

Yaghmai et al.
Imaging Assessment of HCC Response to Therapy

Gastrointestinal Imaging
Review

Imaging Assessment of
Downloaded from www.ajronline.org by 36.76.228.89 on 10/09/17 from IP address 36.76.228.89. Copyright ARRS. For personal use only; all rights reserved

Hepatocellular Carcinoma
FOCUS ON:

Response to Locoregional
and Systemic Therapy
Vahid Yaghmai1 OBJECTIVE. This article focuses on imaging techniques and imaging findings after lo-
Cecilia Besa2 coregional and systemic therapy in patients with hepatocellular carcinoma (HCC). We dis-
Edward Kim 2 cuss currently available locoregional and systemic therapies for HCC, imaging modalities
Joseph L. Gatlin1 and protocols used to assess HCC response, criteria for assessing HCC response, imaging ap-
Nasir A. Siddiqui1 pearances of treated HCC, and future directions.
CONCLUSION. The evaluation of tumor response after systemic and locoregional thera-
Bachir Taouli2
pies is essential in directing management for HCC. An understanding of the various therapeu-
Yaghmai V, Besa C, Kim E, Gatlin JL, Siddiqui NA, tic strategies and of their posttherapy imaging appearances is essential for accurately assessing
Taouli B treatment response. The evaluation of tumor response should include not only anatomic im-
aging biomarkers, such as reduction in tumor size, but also tumor enhancement and necrosis.

H
epatocellular carcinoma (HCC) Locoregional Treatment Options
is the most common primary he- for Hepatocellular Carcinoma
patic malignancy and usually de- Locoregional therapy is an important part of
velops in the setting of liver cir- the management of HCC because tumors are
rhosis. Over the past 20 years, the incidence resectable or meet transplant criteria at the time
of HCC in the United States has increased of diagnosis in only 510% of patients [10]. In
from 1.5 to 4.9 cases per 100,000 individuals, addition, locoregional therapy has the advan-
with a concomitant 41% increase in the over- tages of preservation of hepatic parenchyma
all mortality rate [1]. Radical treatment strat- and overall less morbidity and mortality com-
egies in HCC involve tumor resection or liver pared with resection. The Barcelona Clinic Liv-
transplantation. Unfortunately, many patients er Cancer classification has been used recent-
have advanced-stage disease or multifocal tu- ly as a guideline for therapies directed toward
mors at presentation and, therefore, are not HCC [11]. The most used locoregional therapy
Keywords: ablation, CT, hepatocellular carcinoma, MRI, candidates for radical treatment options [2]. that will be discussed consists of imaging-guid-
transarterial chemoembolization
In these cases, systemic or locoregional ther- ed percutaneous ethanol or thermal ablation,
DOI:10.2214/AJR.13.10706 apy is used to treat the disease with promis- such as radiofrequency ablation (RFA) and
ing results reported in experienced hands [3]. microwave ablation, TACE, and transarterial
Received February 8, 2013; accepted without revision Locoregional therapy such as transarterial radioembolization, and the emerging technol-
February 11, 2013.
chemoembolization (TACE) is also used as a ogy of irreversible electroporation.
1
Department of Radiology, Northwestern University,
bridge to liver transplantation or to down- For early-stage HCC, imaging-guided tumor
Feinberg School of Medicine, Chicago, IL. stage tumors exceeding Milan criteria [48]. ablation has been recommended in patients
Imaging evaluation of HCC response to who are not suitable candidates for resection.
2
Department of Radiology, Icahn School of Medicine at therapy is generally performed with cross- Percutaneous ethanol ablation induces coagu-
Mount Sinai, Translational and Molecular Imaging
sectional imaging (CT and MRI). Accurate lation necrosis through cellular dehydration,
Institute, One Gustave Levy Pl, Box 1234, New York, NY
10029. Address correspondence to B. Taouli assessment of response to therapy, either lo- protein denaturation, and chemical occlusion
(bachir.taouli@mountsinai.org). coregional or systemic, requires evaluation of of small tumor vessels [12]. Ethanol ablation
tumor size, tumor margins, and tumor necrosis is now rarely used in the era of efficient RFA
AJR 2013; 201:8096 as well as early detection of residual or recur- methods because of inhomogeneous injection
0361803X/13/201180
rent tumor and new tumor. The evaluation of and a high recurrence rate in tumors with a di-
treatment success is essential for future treat- ameter of greater than 3 cm [13]. Thermal ab-
American Roentgen Ray Society ment decisions and for prognosis [9]. lation alters tissue temperature to induce cellu-

80 AJR:201, July 2013


Imaging Assessment of HCC Response to Therapy

lar disruption and tissue coagulation necrosis. tration radiation causing tumor destruction Sorafenib, an oral multikinase inhibitor that
RFA has been shown to be an effective thera- (SIR-Spheres, Sirtex; or TheraSpheres, Nor- targets both tumor cell viability and tumor an-
py, with overall survival and disease-free sur- dion) [26]. Studies have shown the efficacy giogenesis, has been shown to improve surviv-
vival similar to resection [14]. In meta-analy- of this therapy to be similar to that of TACE al when compared with placebo [31]. These re-
ses of randomized controlled trials, RFA was and have documented its safety [2729]. sults have established sorafenib monotherapy
shown to have greater effectiveness and sur- Lower toxicity and longer time to progres- as the reference standard for unresectable HCC.
vival than percutaneous ethanol injection [15 sion have been cited as advantages of trans- Variation in efficacy based on the cause of the
Downloaded from www.ajronline.org by 36.76.228.89 on 10/09/17 from IP address 36.76.228.89. Copyright ARRS. For personal use only; all rights reserved

17]. Patients with very-early-stage HCC have arterial radioembolization [30]. HCC has been suggested by preliminary data
complete response rates of 97% with 5-year Advanced-stage HCC, according to the that show patients with chronic hepatitis C have
survival rates of 68% [18]. A factor that lim- Barcelona Clinic Liver Cancer classification, a better response to sorafenib than patients with
its the use of RFA is vessel size of greater than should be treated with sorafenib, a system- HCC from other causes [38]. Additionally, the
3 mm abutting the target tumor, which causes ic multikinase inhibitor, which we discuss in synergistic effects of sorafenib combined with
heat loss due to perfusion-mediated tissue the next section [31]. In patients with difficul- conventional chemotherapeutic agents have
cooling [7]. Microwave ablation has recent- ty tolerating sorafenib, locoregional therapy yet to be defined; however, initial studies have
ly emerged as a viable alternative for thermal may be a viable alternative. In patients with shown that sorafenib combined with doxorubi-
ablation. The advantages of microwave abla- branch vascular invasion, TACE has provided cin has promise in reducing tumor progression
tion over RFA are higher intratumoral tem- an overall survival benefit similar to sorafenib and increasing survival duration [39].
peratures, larger tumor ablation volumes, fast- [32]. In comparison with sorafenib, transarte- Bevacizumab, a molecular targeted mono-
er tumor ablation, and increased resistance to rial radioembolization has also shown similar clonal antibody directed against tumor angio-
tissue cooling due to adjacent vessels [19]. overall survival in patients with segmental and genesis, has shown promise both alone and
Because of the recent technologic advances in main portal vein invasion [33]. Results from in combination with TACE [40]. Combined
microwave ablation, only a small number of randomized controlled trials are ongoing to therapy with TACE offers an intriguing op-
studies regarding its effectiveness have been establish the promising role of transarteri- tion because some of the effects of TACE may
published, but the results are promising. al radioembolization in the Barcelona Clinic be blunted by neoangiogenesis, which would
A novel, nonthermal ablative technology Liver Cancer algorithm. be impeded by the use of bevacizumab [34].
that is undergoing clinical investigation for However, the efficacy of this treatment option
early-stage HCC is irreversible electropora- Systemic Therapy Options for has yet to be verified against standard thera-
tion. Irreversible electroporation induces ir- Hepatocellular Carcinoma pies in randomized controlled trials. Addition-
reversible disruption of cell membrane integ- Medical treatment options have signifi- al molecular targeted agents as well as gene
rity by altering transmembrane potentials, cantly advanced over the past decade because and cell therapies are being pursued with the
resulting in cell death [20]. Because of its the biology and the pathogenesis of HCC tu- hopes of reducing the carcinogenesis and pro-
nonthermal technology, irreversible electro- morigenesis are better understood [34]. How- liferative behavior of HCC [34]. The challenge
poration is not susceptible to the cooling that ever, despite this progress, systemic therapy will be to identify objective imaging biomar-
occurs during thermal ablation of tumors ad- remains reserved for advanced, unresectable kers that accurately assess treatment response.
jacent to vessels. Results of ongoing prospec- HCC in which radical therapy or liver-directed In summary, there are several options for
tive clinical trials are needed. therapy is not possible [35]. Conventional cy- locoregional therapy in HCC, which may in-
For patients with intermediate-stage HCC, totoxic chemotherapy has, for the most part, clude thermal ablation, microwave ablation,
TACE has been established as the standard of failed to provide long-term benefits. The fail- and TACE with or without drug-eluting beads.
care on the basis of pivotal randomized con- ure of chemotherapy has been attributed large- These therapies can be used in synergy with
trolled trials of TACE compared with best ly to tumor cell resistance and the pharmaco- one another depending on the institution.
supportive care [2123]. With the advent of kinetics in the setting of cirrhosis, which alters Sorafenib is the only systemic drug approved
a drug-eluting beads platform for TACE, pa- the side effect profile and tolerability of many for use in unresectable HCC that has shown
tients have decreased systemic side effects nonselective chemotherapy agents [34]. Addi- improved survival compared with placebo.
with significant reductions in serious liver tionally, hormonal agents, such as tamoxifen
toxicity [24, 25]. TACE with drug-eluting or octreotide, have failed to show an objective Imaging Techniques for Assessment
beads allows sustained and controlled deliv- treatment response in placebo-controlled ran- of Hepatocellular Carcinoma
ery of doxorubicin as compared with iodized domized trials or when added to existing che- Response
oil (Lipiodol, Guerbet)based TACE (con- motherapeutic options [36, 37]. Ultrasound and Contrast-Enhanced Ultrasound
ventional TACE). The objective response rate Molecular targeted therapies directed against Ultrasound is generally used for HCC screen-
between conventional TACE and TACE with specific molecular alterations in the patho- ing [41] and is not recommended for follow-
drug-eluting beads was not statistically signifi- physiology of HCC have shown the most up of patients with treated HCC. On the other
cant [24]. Transarterial radioembolization has promise. Unlike conventional chemotherapy, hand, there has been considerable interest in
emerged as a therapeutic option for patients molecular targeted therapies aim to prevent using contrast-enhanced ultrasound (CEUS)
with intermediate-stage HCC. This therapy the growth and spread of disease by interfer- to characterize solid liver masses such as HCC
differs from TACE in that microspheres con- ing in the signaling pathways for tumor pro- [42, 43]. CEUS has also been studied to as-
taining 90Y, a -emitting isotope, are injected gression and viability [34]. These cytostatic sess response to locoregional therapy includ-
into hypervascular HCC through flow-direct- agents are geared toward increased efficacy ing TACE [44], RFA [45], percutaneous etha-
ed therapy in which high-energy, low-pene- and reduced toxicity. nol ablation, and combined techniques [46].

AJR:201, July 2013 81


Yaghmai et al.

On postablation CEUS, nodules showing no may be feasible and has potential dose reduc- techniques with MRI have been shown to be
contrast enhancement in the arterial phase cor- tion advantages [52]. Increased detection of beneficial in depicting residual enhancement,
relate with complete necrosis on CT and nod- hypervascular lesions with DECT has also with excellent correlation with histopatho-
ules with persistent arterial vascularization are been reported [53] and DECT may improve logic degree of tumor necrosis [61]. The add-
considered viable tumor deposits [46]. characterization of indeterminate subcentime- ed value of gadoxetic acid over extracellular
The potential benefits of CEUS include its ter lesions using a generated iodine map [51]. GBCAs after HCC locoregional therapy is un-
ease of use during or immediately after locore- DECT may also have utility in evaluating clear. A recent study suggested no benefit of
Downloaded from www.ajronline.org by 36.76.228.89 on 10/09/17 from IP address 36.76.228.89. Copyright ARRS. For personal use only; all rights reserved

gional therapy and the fact that high-densi- HCC response to locoregional therapy, such using the hepatobiliary phase over dynamic
ty iodized oils used during TACE do not limit as RFA, with higher lesion-to-liver contrast- phases after gadoxetic acid administration for
CEUS interpretation, as can be the case with to-noise ratio on iodine maps, which aids in the detection of local recurrence of HCC after
CT [44]. Some investigators have reported the detection of residual tumor [54]. RFA [62], whereas another study comparing
CEUS to be more sensitive than contrast-en- CT with gadoxetic acidenhanced MRI after
hanced CT (CECT) for the detection of small MRI RFA showed higher interobserver agreement
foci of residual tumor after TACE [44]. How- State-of-the-art protocols using 1.5- or 3-T for MRI than CT and a better performance in
ever, to date, microbubble contrast agents for MRI systems include breath-hold sequences general for MRI [63]. The added potential val-
the evaluation of liver lesions have not re- for routine MRI examination of the liver (ac- ue of gadoxetic acid may be in the detection of
ceived approval by the U.S. Food and Drug quisition time for whole-liver imaging < 30 new lesions and in the differentiation of small
Administration (FDA), and this technique re- minutes). The following sequences are recom- areas of viable tumor from pseudolesions en-
mains rarely performed in the United States. mended: axial gradient-recalled echo (GRE) hancing during the arterial phase by assess-
in phase and opposed phase T1-weighted im- ing washout on the late venous phase or lack
MDCT aging, axial fast spin-echo T2-weighted imag- of functioning hepatocytes (hypointensity) on
CT has long been a mainstay of liver and ing with fat saturation, axial and coronal T2- the hepatobiliary phase [64].
HCC imaging for both initial tumor charac- weighted HASTE imaging, diffusion-weighted
terization and posttreatment follow-up for re- imaging (DWI), and axial 3D fat-suppressed PET
sponse assessment. Biphasic scanning in the GRE T1-weighted imaging before and after FDG PET has limited sensitivity for HCC
hepatic arterial and portal venous phases is gen- dynamic injection of extracellular gadolini- detection ( 60%) and its role in assessing
erally used for HCC detection [4749]. MDCT um-based contrast agents (GBCAs) or a liver- HCC response to therapy needs to be validat-
uses 16, 64, 128, or more contiguous detectors specific GBCA (such as gadoxetate disodium ed [65]. Studies have shown a higher sensi-
to increase effective pitch without consequent [Eovist or Primovist, Bayer HealthCare Phar- tivity of 18F-fluorocholine for detecting HCC,
loss of spatial resolution along the axis of scan- maceuticals]) or gadoxetic acid using an MR- especially well-differentiated HCC, when
ning. These scanners allow thin-section imag- compatible automatic injector. The volumetric compared with FDG [66]. However, the lack
es to be obtained in a single breath-hold with T1-weighted imaging sequence is the most im- of tumor specificity of FDG PET, limited
greatly improved speed and longitudinal res- portant sequence for HCC detection and char- quantitative data on the role of the standard-
olution, which results in nearly isotropic im- acterization; acquisitions are performed at the ized uptake value in predicting overall sur-
age acquisition and subsequent high-resolution hepatic arterial phase using either a test bolus of vival of patients with HCC, and limitations
multiplanar reformations. For patients suspect- contrast material or a bolus-tracking method for in reproducibility of semiquantitative meth-
ed of having HCC, the United Network for Or- accurate timing [55]. The portal venous phase ods in FDG PET support the need for addi-
gan Sharing currently recommends the use of a is at 1 minute after contrast injection late ve- tional studies to establish its role in the man-
quadruple-phase CT protocol that includes un- nous or equilibrium phase, at 3 minutes (for ga- agement of patients with HCC.
enhanced images (to characterize residual en- doxetic acid); and hepatobiliary phase, at 10 or
hancement in posttreatment cases), a single late 20 minutes (for gadoxetic acid) [56, 57]. Con- Summary
arterial phase based on a bolus-tracking meth- trast-enhanced dynamic T1-weighted imaging There is no clear evidence showing the su-
od (for accurate peak arterial enhancement), a with DWI can be advantageous in assessing periority of MRI or CT for assessing HCC re-
portal venous phase at 60 or 70 seconds after treatment-related changes in HCC and has been sponse to therapy. Therefore, the choice of CT
iodine contrast injection at a rate of 45 mL/s, shown to be superior to CECT in evaluating or MRI depends on local expertise and avail-
and a late venous phase ( 120 seconds) after patients who have undergone Lipiodol-based ability. However, we recommend that MRI
iodine contrast injection [50]. TACE therapies [58, 59]. The beam-hardening with subtraction be used for the follow-up of
Recently, dual-energy CT (DECT) has be- effects of high-attenuation Lipiodol on CT may patients treated with Lipiodol-based TACE
come available and its utility in imaging hy- obscure small enhancing tumors. However, and in patients with questionable areas of re-
pervascular liver masses such as HCC is being Lipiodol does not adversely affect MR signal- sidual enhancement on CT after locoregion-
evaluated. DECT provides additional infor- intensity characteristics, so residual enhance- al therapy. Regarding the choice of an ex-
mation about how tissues of differing densi- ment can be detected, especially when image tracellular GBCA or a liver-specific GBCA,
ties behave at differing tube voltages. DECT subtraction is used [58, 59]. because the role of MRI for the diagnosis of
can be performed with either two x-ray sourc- Lesions treated with RFA or TACE typically recurrence relies on the dynamic phases of en-
es of differing tube voltages or a single source undergo coagulative hemorrhagic necrosis that hancement, there is no compelling evidence
with rapidly alternating tube voltages [51]. may appear hyperintense on unenhanced T1- to use a liver-specific agent for follow-up of
The omission of a true unenhanced phase to weighted imaging, making contrast-enhanced treated HCC other than for detecting new tu-
be replaced with a virtual unenhanced phase evaluation difficult [60]. Image subtraction mor foci away from the treated lesion.

82 AJR:201, July 2013


Imaging Assessment of HCC Response to Therapy

TABLE 1: Comparison of European Association for the Study of the Liver (EASL) Criteria [72] and Modified Response
Evaluation Criteria in Solid Tumors (RECIST) [73] for the Assessment of Hepatocellular Carcinoma (HCC)
Response to Locoregional and Systemic Therapy
Assessment Category EASL Criteriaa Modified RECISTb
CR Disappearance of all known disease and no new lesions determined by Disappearance of any intratumoral arterial
two observations not less than 4 weeks apart enhancement in all target lesions
Downloaded from www.ajronline.org by 36.76.228.89 on 10/09/17 from IP address 36.76.228.89. Copyright ARRS. For personal use only; all rights reserved

PR At least 50% reduction in total tumor load of all measurable lesions At least 30% decrease in the sum of diameters of viable
determined by two observations not less than 4 weeks apart target lesions
SD Any cases that do not qualify for either PR or PD Any cases that do not qualify for either PR or PD
PD At least 25% increase in size of one or more measurable lesions or the At least 20% increase in the sum of the diameters of
appearance of new lesions viable target lesions
NoteBoth methods are based on CT or MRI using contrast-enhanced arterial phase images (see also Fig. 1). CR = complete response, PR = partial response, SD =
stable disease, PD = progressive disease.
aBidimensional measurements.
bUnidimensional measurements.

Criteria for Response Assessment in the first few weeks after therapy, because comings of the EASL criteria by defining
in Hepatocellular Carcinoma tumor shrinkage may be delayed. In fact, after methods for newer image acquisitions, target
WHO Guidelines and RECIST locoregional therapy, a treated HCC can possi- selection, and target measurement by adapt-
Conventional oncologic imaging has fo- bly increase in size secondary to intratumoral ing many of the strengths of RECIST. Modi-
cused on anatomic biomarkers to assess tu- edema, hemorrhage, or necrosis [70]. There- fied RECIST uses the single largest diameter
mor response. The World Health Organization fore, better criteria adapted to these new thera- of the viable tumor (defined as the compo-
(WHO) guidelines, incorporating bidimen- pies have been developed. nent enhancing during the arterial phase) and
sional perpendicular measurements [67], and is more practical for clinical use (Table 1 and
the Response Evaluation Criteria in Solid Tu- EASL Criteria and Modified RECIST Fig. 1). The EASL and European Organisa-
mors (RECIST) (Fig. 1), incorporating uni- Given the limitations of the WHO guidelines tion for Research and Treatment of Cancer
dimensional measurements, were intended to and RECIST, new criteria taking into account (EORTC) [74] have recently endorsed the
evaluate change in tumor size over months to a decrease in enhancing tumor and tumor ne- use of the modified RECIST criteria for the as-
years after systemic treatments and do not take crosis on CT and MRI have been proposed [71, sessment of HCC response based on dynamic
into account changes in tumor vascularity or 72]. The criteria proposed by the European As- CT or MRI performed 1 month after locore-
necrosis [68, 69]. However, because the ob- sociation for the Study of the Liver (EASL) are gional therapy or systemic therapy.
jective of effective locoregional therapy is to based on modified WHO bidimensional mea- Several studies have shown modified RECIST
obtain tumor necrosis regardless of the pres- surements to estimate tumor response (Table to be superior to RECIST in predicting HCC
ence of changes in size, response based on size 1 and Fig. 1). response to TACE and antiangiogenic therapy
changes may not be achieved by the new cy- The recently introduced modified RECIST [7577]. Measurements of the two largest tar-
tostatic and locoregional therapies, especially classification [73] addresses many of the short- get lesions have been shown to be adequate for

RECIST EASL Modified RECIST

Fig. 1Diagrams compare different imaging response criteria used in evaluation of hepatocellular carcinoma (HCC) after treatment: Response Evaluation Criteria in
Solid Tumors (RECIST) [68, 69], European Association for the Study of the Liver (EASL) criteria [72], and modified RECIST [73, 74]. A partially necrotic (black) HCC with an
enhancing component (white) is shown. Arrows illustrate measurements obtained for response assessment. RECIST measures single largest dimension of lesion; EASL
measures two dimensions of enhancing viable tumor; modified RECIST measures single largest dimension of enhancing viable tumor. Both EASL criteria and modified
RECIST are based on measurements obtained on CT or MRI using contrast-enhanced arterial phase images.

AJR:201, July 2013 83


Yaghmai et al.
Downloaded from www.ajronline.org by 36.76.228.89 on 10/09/17 from IP address 36.76.228.89. Copyright ARRS. For personal use only; all rights reserved

A B C
Fig. 259-year-old man with cirrhosis from chronic hepatitis C virus infection and hepatocellular carcinoma (HCC) who presented for follow-up imaging after undergoing
microwave ablation.
A, Pretreatment axial arterial phase CT image shows hypervascular HCC (arrow) in right hepatic lobe.
B and C, Immediate postprocedural axial unenhanced (B) and contrast-enhanced (C) arterial phase CT images show central high density (arrowhead, B) related
to hemorrhage and tiny air bubbles (arrowhead, C) within ablation zone and surrounding liver. Air bubbles are expected finding immediately after ablation due to
boiling of tissue.

the assessment of HCC response to TACE when so tumors are treated at different time points, weeks after locoregional therapy or after the
using modified RECIST guidelines [78, 79]. In which decreases the value of these criteria start of systemic therapy. They also recom-
two recent studies of patients with HCC treated when assessing locoregional therapy. mend performing imaging follow-up for the
with TACE, modified RECIST and EASL criteria detection of recurrence and new lesions ev-
independently predicted overall survival [77, 78]. Summary ery 3 months during at least the first 2 years
Response assessments based on EASL criteria The modified RECIST criteria take into ac- after treatment begins. Afterward, ultrasound
and modified RECIST performed approximately count size and vascularity (on arterial phase is recommended every 6 months.
1 month after therapy with TACE using drug-elut- CT or MRI) of residual enhancing tumor and
ing beads have been shown to predict survival, are appropriate for use in HCC treated with lo- Appearance After Locoregional Therapy
with better performance for the modified RECIST coregional therapy. However, there are cases Evaluation during and immediately after
guidelines [76]. In addition, a recent study [80] (e.g., infiltrative tumors, heterogeneous ne- locoregional therapyPeriprocedural evalu-
showed a significant association between overall crosis) and situations (e.g., after ablation) in ation of transcatheter therapies such as TACE or
survival and EASL and modified RECIST re- which the modified RECIST criteria are diffi- transarterial radioembolization can be challeng-
sponses, whereas there was no significant associa- cult or impossible to apply. ing because the target tumor may have a vari-
tion between survival and RECIST 1.1 response. able blood supply. Identification of the vessels
Data on the performance of modified RECIST Imaging Appearances of Treated that perfuse the tumor is essential in directing
in assessing HCC response to systemic thera- Hepatocellular Carcinoma these therapies and ensuring that an adequate
pies are limited. In a study of 53 patients treat- General Imaging Considerations ablative response is achieved with minimal col-
ed with sorafenib, responders based on modi- The ultimate goal of locoregional therapy of lateral damage [84]. Conventional digital sub-
fied RECIST showed longer overall survival HCC is tumor cell death and necrosis [82]. Sys- traction angiography is limited in assessing the
than nonresponders [75]. The combination of temic therapies in HCC are typically cytostatic, variable vascular supply to a tumor and also in
-fetoprotein level and modified RECIST has leading to little change in tumor size. Because determining therapeutic efficacy [85]. In some
been shown to predict prognosis in patients with of the complexities in therapeutic approaches, centers, imaging during locoregional therapy
advanced HCC treated with sorafenib [81]. a therapy-tailored imaging evaluation of HCC may be performed with C-arm cone beam CT.
The modified RECIST criteria have limita- after therapy is of paramount importance [83]. This technique offers reconstructed CT-like im-
tions: They have not been validated for assess- Accurate interpretation of posttherapy changes ages from a flat-panel C-arm unit during the ab-
ing HCC after transarterial radioembolization in tumor by evaluating lesion margins and vi- lative therapy, thereby allowing visualization of
and RFA. Also, it is difficult to measure with ability is essential for assessing response and the soft-tissue tumor and feeding vessels [84].
confidence diffusely necrotic lesions with inter- determining the need for future treatment [64]. Additionally, the distribution of embolic ma-
vening viable components. Finally, many pa- The EASL-EORTC [74] guidelines rec- terials such as Lipiodol can be evaluated imme-
tients with multiple HCCs are treated in stages, ommend performing dynamic CT or MRI 4 diately or within 24 hours after the procedure

84 AJR:201, July 2013


Imaging Assessment of HCC Response to Therapy

[86]. However, these practices vary from center lation site (Fig. 3). This coagulative necrosis of residual tumor may be obscured by tran-
to center, are beyond the scope of this article, will result in a hyperdensity within the treated sient hyperemia, so cases showing persistent
and will not be discussed here. HCC on CT and hyperintense signal on T1- arterial enhancement with washout on delayed
For ablation techniques, success is defined weighted imaging, thus limiting assessment phase images at short-term follow-up should
as a nonenhancing ablation zone encompassing for residual tumor enhancement. However, be evaluated further to determine whether ad-
the entire tumor and an ablative safety margin these findings frequently will have resolved on ditional directed therapy is needed [82].
of at least 5 mm of the nontumor hepatic paren- subsequent imaging [89] (Fig. 2). Subtraction Regardless of the mode of therapy, nodular
Downloaded from www.ajronline.org by 36.76.228.89 on 10/09/17 from IP address 36.76.228.89. Copyright ARRS. For personal use only; all rights reserved

chyma [87]. Moreover, tiny air bubbles that re- images can be a helpful adjunct for differen- or thick areas of arterial enhancement along
sult from the boiling of tissue can be seen im- tiating hemorrhage from enhancing tumor on the margin of a treated HCC are consistent
mediately after RFA and will typically have MRI [61] (Figs. 4 and 5). Transient hyperemia with residual tumor, especially when there
resolved by 1-month follow-up [88] (Fig. 2). manifested by thin, uniform enhancement of is associated washout, and require additional
The air bubbles should be differentiated from the treated zone is an expected finding after treatment [60, 88] (Fig. 6). In a recent retro-
the mottled persistent air densities seen with a RFA, TACE, or transarterial radioemboliza- spective study, a lack of tumor enhancement,
hepatic abscess or within a peripheral wedge- tion and represents a transient physiologic re- peripheral ring enhancement, and peripheral
shaped defect of a hepatic infarct [88]. How- sponse to thermal injury and embolization to nodular enhancement on arterial phase CT
ever, we will not describe complications of lo- the hepatic parenchyma [82] (Figs. 4 and 5). after TACE with drug-eluting beads showed
coregional therapy in this article. This finding should be differentiated from re- progressively greater correlation with tumor
Follow-up evaluation after locoregional sidual malignancy [89]. Typically transient en- progression (i.e., 8%, 17%, and 83%, re-
therapyA central area of coagulative necro- hancement resolves within several months on spectively) [90]. However, in cases with in-
sis is often seen within the tumor or the ab- follow-up imaging [89]. However, small foci conclusive imaging findings, short-term fol-

TACE or 1. Ablation 1.
transarterial
radioembolization HCC HCC

CR PR or recurrence CR PR or recurrence

2. 3. 2. 3.

* *

4. 4. 5.

Tumor shrinkage Cavity shrinkage

A B
Fig. 3Diagrams illustrate changes in hepatocellular carcinoma (HCC) after treatment. CR = complete response, PR = partial response.
A, Treatment changes after transarterial chemoembolization (TACE) or transarterial radioembolization. Hypervascular HCC before treatment is illustrated as white circle
in diagram 1. Diagrams 2 and 4 shown below CR label illustrate appearances of successful locoregional therapy: lack of enhancement of treated lesion compatible with
complete necrosis (black circle, diagram 2) with possible surrounding geographic arterial enhancement, rim enhancement, or both (asterisk, diagram 2) corresponding to
posttreatment changes. Enhancement may be segmental or lobar for transarterial radioembolization and rim enhancement surrounding lesion is possible, especially after
transarterial radioembolization. Soon after TACE or transarterial radioembolization, treated lesion shows no changes in size but can decrease in size over time (diagram
4); delayed shrinkage may be seen after transarterial radioembolization. Diagram 3 under PR or recurrence label shows residual enhancing tumor.
B, Treatment changes after ablation. Hypervascular HCC before treatment is illustrated as white circle in diagram 1. Diagrams 2 and 4 below CR label show ablation
cavity (double-headed arrow) is larger than original tumor because of ablation margin. Surrounding geographic arterial enhancement, rim enhancement, or both (asterisk,
diagram 2) corresponding to posttreatment changes is shown. Diagrams 3 and 5 under PR or recurrence label show small nodule that increases in size on follow-up
imaging.

AJR:201, July 2013 85


Yaghmai et al.
Downloaded from www.ajronline.org by 36.76.228.89 on 10/09/17 from IP address 36.76.228.89. Copyright ARRS. For personal use only; all rights reserved

Fig. 456-year-old man with cirrhosis from chronic hepatitic C virus infection and hepatocellular carcinoma (HCC). Axial fat-suppressed MR images obtained before
(AD) and 7 months after (EH) transarterial chemoembolization (TACE) with drug-eluting beads and stereotactic radiation therapy are shown.
A, T2-weighted image obtained before therapy shows 3.8-cm hyperintense HCC (arrow) in segment VIII (see also Fig. 13).
B, Unenhanced 3D gradient-recalled echo (GRE) T1-weighted image shows that HCC (arrow) is hypointense.
C and D, Three-dimensional GRE T1-weighted images obtained during arterial phase (C) and portal venous phase after injection of extracellular gadolinium-based
contrast agent (D) show HCC (arrow) exhibiting wash-in and washout and that capsule is visible.
E, T2-weighted image obtained 7 months after therapy shows that HCC (white arrow) is hypointense and smaller (2.4 cm). There is also capsular retraction (black arrow).
F, Unenhanced 3D GRE T1-weighted image shows increased signal intensity in treated HCC (arrow).
G, Subtracted arterial phase 3D GRE T1-weighted image after injection of gadoxetic acid shows HCC (long arrow) is completely necrotic with geographic arterial
enhancement of surrounding liver parenchyma (short arrows), which became isointense to liver parenchyma, compatible with perfusion abnormalities.
H, Three-dimensional GRE T1-weighted image obtained at hepatobiliary phase shows hypointense necrotic lesion (long arrow) and decreased uptake of surrounding liver
parenchyma (short arrows); these findings are likely secondary to external beam radiation therapy and TACE.

low-up in 3 months may be necessary to assess nodule [95] (Figs. 6 and 8). Hyperintense ap- ue and specificity for the prediction of necro-
the resolution or progression of residual dis- pearance on T2-weighted imaging or DWI sis and is not necessarily indicative of viable
ease in the treated lesion or cavity. with associated nodular enhancement is char- tumor [98]. It is also important to emphasize
Next, we discuss some findings specific to acteristic of tumor or incompletely treated tu- the delayed effect of transarterial radioembo-
the locoregional therapy used. After TACE mor (Figs. 6 and 8). Care should be taken not lization on tumor necrosis and shrinkage, with
using Lipiodol, unenhanced CT could be per- to diagnose peripheral regrowth when a thin, reported median times to response of approx-
formed to assess Lipiodol distribution. In regular rim of progressive enhancement is imately 30 and 120 days, using necrosis and
general, complete retention of iodized oil has present because this finding is a sign of simple size criteria, respectively [100] (Fig. 10).
high correlation with complete lesion necro- vascularized inflammation or fibrosis [96]. In In summary, when interpreting the first post-
sis and incomplete retention corresponds to addition, after RFA, arterioportal shunts can procedural imaging study, usually obtained
a range of responses from complete necro- be seen due to thermal injury to small vessels 4 weeks after the procedure, it is important to
sis to residual viable tumor [91, 92] (Fig. 7). in the hepatic parenchyma but will resolve on differentiate normal posttreatment appearance
Lack of iodized oil uptake immediately after follow-up evaluation [97]. (shunting surrounding the lesion, thin rim of
therapy may also indicate an aberrant vascu- After transarterial radioembolization, the enhancement, T1 hyperintense or hyperdense
lar supply to the tumor and will require re- radiation effect is manifested as heterogene- cavity or treated tumor, heterogeneous per-
peat treatment as soon as possible [93, 94]. ous enhancement of the liver in a perivascu- itumoral enhancement) from residual tumor
However, multiphasic MDCT must still be lar distribution in a treated segment or lobe. and recurrent tumor (nodule with wash-in and
performed to assess areas lacking iodized oil This posttherapy finding is unique to trans- washout, with or without T2 hyperintensity,
[91]. Necrotic nonviable tumor tissue will arterial radioembolization and could be eas- with or without diffusion restriction).
continue to retain the oil indefinitely but will ily mistaken for tumor progression [98, 99].
decrease in size over time. However, it is An explant correlation study assessing HCC Appearance After Systemic Therapy
preferable to perform an MRI study because after transarterial radioembolization showed At MDCT, sorafenib has been shown to de-
MRI is more accurate for assessing residual that decreased enhancement and size of treat- crease HCC enhancement without necessarily
or recurrent tumor than CT [59]. ed lesions correlate with pathologic necrosis, affecting tumor size [101]. The predominant
After ablation, residual viable tumor or tu- whereas peripheral nodular enhancement indi- patterns of HCC response after sorafenib on
mor regrowth tends to be detected at the pe- cates the presence of viable tumor (Fig. 9). The MRI are T1 and T2 tumor hyperintensity and
riphery of the treated cavity either as irregu- same study showed that the presence of rim necrosis [102] (Fig. 11). An increase in tumor
lar thickening at the margin or as a new tumor enhancement had 93% positive predictive val- size due to necrosis has also been reported.

86 AJR:201, July 2013


Imaging Assessment of HCC Response to Therapy

Therefore, quantification of necrosis appears


to be a better method of treatment response
assessment than tumor size in HCC treated
with multikinase inhibitors [102]. A recent pi-
lot study showed a statistically significant im-
provement in time to progression of disease
in patients with advanced HCC treated with
Downloaded from www.ajronline.org by 36.76.228.89 on 10/09/17 from IP address 36.76.228.89. Copyright ARRS. For personal use only; all rights reserved

sunitinib when tumor density decreased 15%


or more [103]. After systemic therapy, simi-
lar to after locoregional therapy, residual thick
and nodular foci of arterial enhancement with-
in HCC suggest residual nontreated tumor.
In summary, even without a decrease in
tumor size, a decrease in enhancement or the
presence of tumor necrosis is an indicator of A B
treatment response after systemic therapy.

Novel Imaging Biomarkers


of Response
Volumetric Evaluation
The anatomic imaging biomarkers assume
that tumors are spherical before and after treat-
ment [104]. In both RECIST and modified
RECIST, a 30% decrease in diameter of tu-
mor, defined as the threshold for partial re-
sponse, is presumed to correspond to a 65%
decrease in tumor volume. Similarly, a 20%
increase in diameter of viable tumor, which
defines the threshold for defining disease pro-
gression, corresponds to an approximately 73%
increase in spherical volume. These cut points C D
are rather arbitrary and may not be applicable
Fig. 561-year-old man with cirrhosis from chronic hepatitic C virus infection and hepatocellular carcinoma
to all therapies. Furthermore, both RECIST and (HCC) after transarterial chemoembolization (TACE) and radiofrequency ablation (RFA) treatments.
modified RECIST measurements are only es- A, Axial unenhanced fat-suppressed 3D gradient-recalled echo (GRE) T1-weighted image obtained 8 months after
timates of the tumor volume and are prone to TACE and RFA shows hyperintense treatment cavity (arrow) in right hepatic lobe; this finding is compatible with
interobserver measurement variability. In a ret- posttreatment hemorrhage.
B, On contrast-enhanced fat-suppressed 3D GRE T1-weighted image obtained at arterial phase after injection of
rospective study of 45 HCCs, diameter based gadoxetic acid, it is difficult to detect any residual enhancement in cavity (arrow).
on 3D measurements was significantly differ- C, Subtracted 3D GRE T1-weighted image obtained at arterial phase shows no evidence of enhancement (long
ent than diameter based on conventional 2D arrow); this finding is compatible with completely necrotic tumor. Peripheral geographic enhancement (short
arrows) is noted adjacent to treatment cavity.
measurements [105]. Similarly, in a study D, Axial fat-suppressed fast spin-echo T2-weighted image shows heterogeneous signal intensity, with areas of hypo-
of 29 HCCs treated with transarterial radio- and hyperintensity, in treated cavity (long arrow) and adjacent geographic high T2 signal intensity (short arrows).
embolization, a significant difference in the
mean calculated necrosis proportions based In summary, volumetric evaluation of HCC Diffusion-weighted imagingDWI has re-
on 2D and volumetric methods was observed and its degree of necrosis is a very promis- cently shown potential for HCC detection (Fig.
[105]. Volumetric evaluation of HCC and its ing tool because it is more accurate and repro- 13) compared to or combined to contrast-en-
necrotic component eliminates this limitation ducible than the currently used bidimensional hanced T1-weighted imaging [109, 110]. DWI
and, when available, offers the most compre- measurement. However, volumetric measure- is also increasingly used to evaluate tumor re-
hensive anatomic evaluation for determining ment is not easily feasible in the routine clin- sponse to chemotherapy, radiotherapy, and lo-
treatment response [106]. Voxel-by-voxel vol- ical setting and is still not included in tumor coregional therapy [111, 112]. There are sev-
umetric analysis of tumor density and necrosis response criteria. eral reports about the use of DWI to evaluate
has been shown to be more reproducible than HCC response to TACE and transarterial ra-
2D analysis [105, 107] (Fig. 12). Volumetric Functional Imaging dioembolization [58, 113117]. These studies
quantification is particularly helpful in cas- Functional imaging, unlike anatomic im- have shown differences in apparent diffusion
es in which necrosis is heterogeneously dis- aging, provides information on tumor viabil- coefficient (ADC) values between viable and
tributed in HCC and cannot be assessed using ity, cellularity, vascularity, and metabolism necrotic portions of HCCs after treatment and
modified RECIST. However, to date, volum- [108]. These changes can be detected earlier measurable differences before and after treat-
etry has not been incorporated into RECIST than anatomic changes and are more applica- ment (Fig. 13). In a prospective study, Kamel
or other commonly used size-based response ble in assessing treatment response after lo- et al. [117] observed an increase in tumor ADC
criteria in HCC. coregional therapy or systemic therapy. value that was significant 12 weeks after ini-

AJR:201, July 2013 87


Yaghmai et al.
Downloaded from www.ajronline.org by 36.76.228.89 on 10/09/17 from IP address 36.76.228.89. Copyright ARRS. For personal use only; all rights reserved

Fig. 639-year-old man with cirrhosis from chronic hepatitic B virus infection and hepatocellular carcinoma (HCC). MR images obtained 2 months after first cycle (AF)
and 2 months after second cycle (GI) of transarterial chemoembolization (TACE) with drug-eluting beads are shown.
A, Axial fat-suppressed T2-weighted image shows 3.4-cm HCC in segment IV with areas of high signal intensity (short arrow) and intermediate signal (long arrow).
B and C, Single-shot echo-planar respiratory-triggered diffusion image obtained using b value of 1000 s/m2 (B) and apparent diffusion coefficient (ADC) map obtained
using b values of 0, 50, 500, and 1000 s/m2 (C) show right-sided nodule (short arrow) to be strongly hyperintense with very low ADC corresponding to coagulative and
hemorrhagic necrosis, whereas viable tumor (long arrow) is less hyperintense with ADC isointense to liver parenchyma.
DF, Three-dimensional gradient-recalled echo (GRE) T1-weighted images (unenhanced, D; contrast-enhanced arterial phase, E; hepatobiliary phase after gadoxetic
acid injection, F) show enhancement of left-sided nodule (long arrow), with lack of contrast retention on hepatobiliary phase compatible with viable tumor, and lack of
enhancement in right-sided nodule (short arrow) compatible with necrosis.
G, Axial fat-suppressed T2-weighted image shows decreased size of treated HCC (arrow) with T2 hypointensity and minimal residual focus of hyperintensity.
H, Axial unenhanced fat-suppressed 3D GRE T1-weighted image shows increased signal intensity in lesion (arrow).
I, Axial contrast-enhanced fat-suppressed 3D GRE T1-weighted arterial phase subtracted image obtained after gadoxetic acid injection shows lack of enhancement in
lesion (arrow); this finding is compatible with complete tumor necrosis.

tial TACE, borderline significant 3 weeks af- was present 12 weeks after TACE. Thus, they gators observed that ADC had a significant cor-
ter therapy, and insignificant 24 hours and 4 recommend the use of contrast-enhanced T1- relation with tumor necrosis assessed with his-
weeks after therapy. They also showed that the weighted imaging and DWI 12 weeks after topathology [118]. For prediction of complete
maximum difference in tumor enhancement TACE. In an explant correlation study, investi- tumor necrosis after TACE, an area under the

88 AJR:201, July 2013


Imaging Assessment of HCC Response to Therapy
Downloaded from www.ajronline.org by 36.76.228.89 on 10/09/17 from IP address 36.76.228.89. Copyright ARRS. For personal use only; all rights reserved

A B C
Fig. 763-year-old man with cirrhosis from chronic hepatitic C virus infection and hepatocellular carcinoma.
A, Pretreatment axial arterial phase CT image shows hypervascular HCC (arrow) in segment IV.
B, Axial unenhanced CT image obtained 24 hours after transarterial chemoembolization (TACE) shows homogeneous dense accumulation of iodized oil (Lipiodol,
Guerbet) in HCC (arrow) and less dense accumulation of Lipiodol in adjacent parenchyma.
C, Axial contrast-enhanced CT image at arterial phase obtained 1 month after TACE shows continued Lipiodol retention in HCC (arrow) while surrounding parenchyma no
longer retains Lipiodol. Lesion has significantly decreased in size, which is compatible with response to therapy. However, enhancement is difficult to assess on CT after
Lipiodol injection.

A B C

D E F
Fig. 867-year-old man who had undergone right hepatectomy for hepatocellular carcinoma (HCC), with recurrent HCC treated with microwave ablation. Follow-up MR
images obtained 2 months after microwave ablation of caudate lobe HCC are shown. DF were obtained slightly superior to AC during same examination.
A, Unenhanced 3D gradient-recalled echo (GRE) T1-weighted image shows treatment cavity (arrow) in caudate lobe with T1 hyperintense rim.
B, Three-dimensional GRE T1-weighted image obtained at arterial phase after injection of gadoxetic acid shows no enhancement (arrow).
C, Subtracted image confirms finding of no enhancement (arrow) seen in B.
D and E, Unenhanced 3D GRE T1-weighted image (D) and arterial phase 3D GRE T1-weighted image after injection of gadoxetic acid (E) show enhancing lesion (arrow) in
caudate lobe adjacent to treatment cavity; this finding is compatible with recurrent HCC.
F, Single-shot echo-planar diffusion image (b value = 1000 s/m 2 ) shows hyperintense nodule (arrow) compatible with restricted diffusion corresponding to recurrent
tumor.

AJR:201, July 2013 89


Yaghmai et al.

Fig. 972-year-old man with hepatocellular


carcinoma (HCC) treated with 90Y radioembolization
with good response. Axial fat-suppressed 3D
gradient-recalled echo T1-weighted images obtained
before and after treatment are shown.
A, Pretreatment contrast-enhanced image at
portal venous phase after injection of extracellular
gadolinium chelates shows HCC (arrow) in segment
VIII.
Downloaded from www.ajronline.org by 36.76.228.89 on 10/09/17 from IP address 36.76.228.89. Copyright ARRS. For personal use only; all rights reserved

B, Three-month follow-up contrast-enhanced image


at late venous phase after injection of extracellular
gadolinium chelates shows complete tumor necrosis.
Central high signal intensity(arrowhead) was also
present on unenhanced T1-weighted image (not
shown) and represents coagulative necrosis. Note
right pleural effusion.
C and D, Six-month (C) and 18-month (D) follow-up
images at portal venous phase after injection of
extracellular gadolinium chelates show persistent
lack of enhancement and progressive decrease in
A B size of HCC. These findings are compatible with
response to therapy.

curve (AUC) of 0.85 was observed for ADC


compared with an AUC of 0.820.89 for im-
age subtraction, without significant difference
between the two techniques [118]. The use of
DWI in combination with conventional MRI
shows promising results in increasing the sen-
sitivity for detecting viable tumor [58] (Figs.
6 and 8). Diffusion restriction (hyperintensity
on imaging performed with high b values and
low ADC values) suggests viable tumor com-
ponents [118, 119]. However, a study showed
lower performance of DWI compared with
contrast-enhanced imaging, with lower sen-
sitivity for detection of local HCC recurrence
C D (60.7% vs 82%, respectively) [120]. Regard-
ing the use of pretreatment ADC as a marker
of response to TACE, the data are limited, and
two studies published to date report conflict-
ing results [121, 122]. In a prospective study by
Yuan et al. [121], nonresponding HCCs had
a significantly higher pretreatment ADC than
HCCs that responded. On the other hand, a re-
cently published retrospective study showed
that HCCs with poor or incomplete response
to TACE had significantly lower pretreatment
ADC and lower post-TACE ADC values than
HCCs with good or complete response [122].
A B
Fig. 1062-year-old man with unresectable
hepatocellular carcinoma (HCC) treated with 90Y
radioembolization with good response.
A, Pretreatment axial arterial phase CT image shows
large enhancing mass (arrow) in left hepatic lobe.
B, Axial posttreatment arterial phase CT image
obtained 1 month after procedure shows progressive
decrease in tumor size and persistent enhancement
(asterisk).
C and D, Follow-up CT images obtained at 3 months
(C) and 12 months (D) after procedure show
continued decrease in size of HCC and capsular
retraction (D). Patient underwent no further therapy.
Response to procedure was good despite areas of
persistent enhancement on early follow-up scans.
C D

90 AJR:201, July 2013


Imaging Assessment of HCC Response to Therapy
Downloaded from www.ajronline.org by 36.76.228.89 on 10/09/17 from IP address 36.76.228.89. Copyright ARRS. For personal use only; all rights reserved

A B C
Fig. 1175-year-old woman with multifocal metastatic hepatocellular carcinoma (HCC) treated with sorafenib and 90Y radioembolization.
A and B, Pretreatment MRI. Axial fat-suppressed 3D gradient-recalled echo (GRE) T1-weighted images at portal venous phase after injection of extracellular gadolinium
chelates show two HCCs (arrow, A; and short arrow, B) in right hepatic lobe. There is large extrahepatic metastatic mass (asterisk) and lung metastasis (long arrow, B).
C, MR image obtained after sorafenib therapy and 90Y radioembolization shows that segment VII lesion (long white arrow) has decreased significantly in size and segment
VIII lesion (short white arrow) has become necrotic but that there has been interval progression of lung metastasis (black arrow). Asterisk = extrahepatic metastatic
mass.

Fig. 1264-year-old man with hepatocellular


carcinoma (HCC) treated with 90Y radioembolization.
A and B, Pretreatment axial contrast-enhanced
CT (CECT) images show enhancing HCC (arrow, A;
orange line, B) in left hepatic lobe. HCC measures 40.5
mm in maximum dimension. Corresponding volume is
15.5 mL after semiautomated segmentation (B).
C and D, Axial CECT images obtained 40 days after
90 Y radioembolization show interval tumor necrosis

(red, D). Maximum size of tumor (arrow, C; orange line,


D) is 33.9 mm. In this case, decrease of 16.2% should
be considered stable disease according to Response
Evaluation Criteria in Solid Tumors. However,
tumor volume has decreased to 8.66 mL (44.1%)
and residual enhancing component (viable tumor)
constitutes only 7.7% of tumor.

diagnosis of tumor recurrence, whereas mea-


A B suring the ADC of the whole ablation zone
was not helpful [123].
The limitations of DWI relate to image qual-
ity, with possible echo-planar imagingrelated
artifacts, and to limited knowledge on ADC
reproducibility in liver tumors [124, 125].
In summary, despite promising results, DWI
cannot replace contrast-enhanced T1-weighted
imaging and subtraction for assessment of HCC
response. The role of baseline ADC and early
changes in ADC values as markers of tumor
response and time to tumor progression should
be determined in a large prospective study.
Perfusion imagingDynamic contrast-en-
hanced MRI (DCE-MRI) [126128] and perfu-
sion CT [129, 130] involve the use of contrast
C D agents with high-temporal-resolution imag-
ing to capture changes in MR signal intensity
Both studies showed an increase in ADC in Regarding the changes in ADC values af- or CT attenuation as a function of time; these
HCC with good response compared with HCC ter ablation, the data are limited. A prior study changes are used to quantify tissue and tumor
with poor response [121, 122]. Given the con- assessed the role of DWI after RFA of liver vascular characteristics. Perfusion CT has the
flicting results from these two studies, the val- tumors including HCCs; the results suggested advantage of a direct linear relation between
ue of pre-TACE ADC in predicting response that measuring ADC in focal hyperintense ar- enhancement change and iodine concentra-
should be verified in a large prospective study. eas inside the ablation zone had value for the tion, whereas the relationship between MR sig-

AJR:201, July 2013 91


Yaghmai et al.

have not been completely validated for routine


clinical use. It is not yet known whether DWI
or DCE-MRI can be used prospectively for the
prediction of subsequent response when used
at baseline and for the early detection of re-
sponse before changes or lack of changes in
size occur. This will help individualize ther-
Downloaded from www.ajronline.org by 36.76.228.89 on 10/09/17 from IP address 36.76.228.89. Copyright ARRS. For personal use only; all rights reserved

apy depending on different tumor profiles.


However, given the lack of standardization
and evidence for utilizing these novel imag-
A B ing methods, morphologic criteria using RE-
CIST or modified RECIST should still be used
in clinical practice and for drug trials.

References
1. El-Serag HB. Epidemiology of hepatocellular
carcinoma in USA. Hepatol Res 2007; 37(suppl
2):S88S94
2. Shields A, Reddy KR. Hepatocellular carcino-
ma: current treatment strategies. Curr Treat Op-
tions Gastroenterol 2005; 8:457466
3. Shanbhogue AK, Karnad AB, Prasad SR. Tumor
C D response evaluation in oncology: current update.
J Comput Assist Tomogr 2010; 34:479484
Fig. 1356-year-old man (same patient as Fig. 4) with cirrhosis from chronic hepatitis C virus infection and
4. Bruix J, Sala M, Llovet JM. Chemoembolization
hepatocellular carcinoma (HCC). Patient underwent transarterial chemoembolization (TACE) with drug-eluting
beads and stereotactic radiation therapy. for hepatocellular carcinoma. Gastroenterology
A, Pretreatment MRI. Axial fat-suppressed single-shot echo-planar diffusion image (b value = 1000 s/mm2) 2004; 127(5 suppl 1):S179S188
shows tumor with restricted diffusion (arrow) corresponding to viable HCC. 5. Yao FY, Hirose R, LaBerge JM, et al. A prospec-
B, Pretreatment apparent diffusion coefficient (ADC) map. Tumor (arrow) ADC was 1.37 10 3 mm2 /s.
C, Follow-up MRI performed 7 months after TACE with drug-eluting beads. Axial fat-suppressed single-shot tive study on downstaging of hepatocellular car-
echo-planar diffusion image (b value = 1000 s/mm2) shows interval resolution of tumor hyperintensity (long cinoma prior to liver transplantation. Liver
arrow) compatible with tumor necrosis and interval decrease in tumor size as well as surrounding hyperintense Transpl 2005; 11:15051514
edema (short arrows) secondary to treatment.
6. Llovet JM, Mas X, Aponte JJ, et al. Cost effec-
D, Follow-up ADC map obtained 7 months after TACE with drug-eluting beads. Tumor (arrow) ADC has
increased to 1.7 10 3 mm2 /s. tiveness of adjuvant therapy for hepatocellular
carcinoma during the waiting list for liver trans-
plantation. Gut 2002; 50:123128
nal intensity and gadolinium concentration is rial fraction and perfusion in tumors effective- 7. Lu DS, Yu NC, Raman SS, et al. Radiofrequency
not necessarily linear depending on the dose ly treated by TACE or RFA. ablation of hepatocellular carcinoma: treatment
of contrast injected, sequence parameters, and In summary, as with DWI, it will be inter- success as defined by histologic examination of
concentration reached in the target tissue. Al- esting to determine the role of pretreatment and the explanted liver. Radiology 2005; 234:954960
though linearity can be assumed in DCE-MRI early changes in tumor perfusion parameters 8. Yao FY, Kerlan RK Jr, Hirose R, et al. Excellent
at certain concentrations, it is preferable to de- as predictors of subsequent response to thera- outcome following down-staging of hepatocellu-
termine gadolinium concentration using unen- py and time to tumor progression. In addition, lar carcinoma prior to liver transplantation: an in-
hanced baseline T1 measurement. In contrast, clinical utility, reproducibility, accuracy, proper tention-to-treat analysis. Hepatology 2008; 48:
perfusion CT is limited by the risk of radiation modeling, and validation of perfusion CT and 819827
exposure, especially when follow-up studies MRI techniques must be established. 9. Mendizabal M, Reddy KR. Current manage-
are needed, and the lack of multiparametric ment of hepatocellular carcinoma. Med Clin
imaging. Multiparametric imaging is possi- Conclusion North Am 2009; 93:885900, viii
ble only with MRI in which DCE-MRI can be The evaluation of tumor response after sys- 10. Llovet JM. Treatment of hepatocellular carcino-
combined with DWI. temic and locoregional therapies is essential ma. Curr Treat Options Gastroenterol 2004;
Recent studies using perfusion CT or DCE- in directing management for HCC. An un- 7:431441
MRI have shown potential for quantifying per- derstanding of the various therapeutic strate- 11. Forner A, Llovet JM, Bruix J. Hepatocellular
fusion of malignant liver lesions and for mon- gies and their posttherapy imaging appearance carcinoma. Lancet 2012; 379:12451255
itoring treatment response to antiangiogenic is essential for accurately assessing treatment 12. Khan KN, Yatsuhashi H, Yamasaki K, et al. Pro-
drugs in liver metastases and HCC [131143]. response. The evaluation of tumor response spective analysis of risk factors for early intrahe-
Antiangiogenic agents are thought to induce should include not only anatomic imaging bio- patic recurrence of hepatocellular carcinoma
an antipermeability effect while TACE reduc- markers, such as reduction in tumor size, but following ethanol injection. J Hepatol 2000;
es tumor blood volume [144]. These effects also tumor enhancement and necrosis. Func- 32:269278
result in a significant decrease in hepatic arte- tional imaging methods are promising but 13. Germani G, Pleguezuelo M, Gurusamy K, Mey-

92 AJR:201, July 2013


Imaging Assessment of HCC Response to Therapy

er T, Isgro G, Burroughs AK. Clinical outcomes drug eluting beads: efficacy and doxorubicin randomized trial. JAMA 2010; 304:21542160
of radiofrequency ablation, percutaneous alco- pharmacokinetics. J Hepatol 2007; 46:474481 40. Siegel AB, Cohen EI, Ocean A, et al. Phase II
hol and acetic acid injection for hepatocellular 26. Atassi B, Bangash AK, Bahrani A, et al. Multi- trial evaluating the clinical and biologic effects
carcinoma: a meta-analysis. J Hepatol 2010; modality imaging following 90Y radioemboliza- of bevacizumab in unresectable hepatocellular
52:380388 tion: a comprehensive review and pictorial essay. carcinoma. J Clin Oncol 2008; 26:29922998
14. Chen MS, Li JQ, Zheng Y, et al. A prospective RadioGraphics 2008; 28:8199 41. Bruix J, Sherman M. Management of hepatocel-
randomized trial comparing percutaneous local 27. Geschwind JF, Salem R, Carr BI, et al. Yttrium-90 lular carcinoma: an update. Hepatology 2011;
Downloaded from www.ajronline.org by 36.76.228.89 on 10/09/17 from IP address 36.76.228.89. Copyright ARRS. For personal use only; all rights reserved

ablative therapy and partial hepatectomy for microspheres for the treatment of hepatocellular car- 53:10201022
small hepatocellular carcinoma. Ann Surg 2006; cinoma. Gastroenterology 2004; 127:S194S205 42. Wilson SR, Burns PN, Muradali D, Wilson JA,
243:321328 28. Salem R, Lewandowski RJ, Atassi B, et al. Lai X. Harmonic hepatic US with microbubble
15. Cho YK, Kim JK, Kim MY, Rhim H, Han JK. Treatment of unresectable hepatocellular carci- contrast agent: initial experience showing im-
Systematic review of randomized trials for hepa- noma with use of 90Y microspheres (Thera- proved characterization of hemangioma, hepato-
tocellular carcinoma treated with percutaneous Sphere): safety, tumor response, and survival. J cellular carcinoma, and metastasis. Radiology
ablation therapies. Hepatology 2009; 49:453459 Vasc Interv Radiol 2005; 16:16271639 2000; 215:153161
16. Crocetti L, de Baere T, Lencioni R. Quality im- 29. Sangro B, Bilbao JI, Boan J, et al. Radioemboli- 43. Youk JH, Kim CS, Lee JM. Contrast-enhanced
provement guidelines for radiofrequency abla- zation using 90Y-resin microspheres for patients agent detection imaging: value in the character-
tion of liver tumours. Cardiovasc Intervent Ra- with advanced hepatocellular carcinoma. Int J ization of focal hepatic lesions. J Ultrasound
diol 2010; 33:1117 Radiat Oncol Biol Phys 2006; 66:792800 Med 2003; 22:897910
17. Orlando A, Leandro G, Olivo M, Andriulli A, 30. Salem R, Lewandowski RJ, Kulik L, et al. Radio- 44. Kim HJ, Kim TK, Kim PN, et al. Assessment of
Cottone M. Radiofrequency thermal ablation vs. embolization results in longer time-to-progression the therapeutic response of hepatocellular carci-
percutaneous ethanol injection for small hepato- and reduced toxicity compared with chemoem- noma treated with transcatheter arterial chemo-
cellular carcinoma in cirrhosis: meta-analysis of bolization in patients with hepatocellular carci- embolization: comparison of contrast-enhanced
randomized controlled trials. Am J Gastroen- noma. Gastroenterology 2011; 140:497507 sonography and 3-phase computed tomography.
terol 2009; 104:514524 31. Llovet JM, Ricci S, Mazzaferro V, et al. J Ultrasound Med 2006; 25:477486
18. Livraghi T, Meloni F, Di Stasi M, et al. Sustained Sorafenib in advanced hepatocellular carcino- 45. Chen MH, Wu W, Yang W, et al. The use of con-
complete response and complications rates after ma. N Engl J Med 2008; 359:378390 trast-enhanced ultrasonography in the selection of
radiofrequency ablation of very early hepatocel- 32. Luo J, Guo RP, Lai EC, et al. Transarterial che- patients with hepatocellular carcinoma for radio
lular carcinoma in cirrhosis: is resection still the moembolization for unresectable hepatocellular frequency ablation therapy. J Ultrasound Med
treatment of choice? Hepatology 2008; 47:8289 carcinoma with portal vein tumor thrombosis: a 2007; 26:10551063
19. Yu NC, Raman SS, Kim YJ, Lassman C, Chang prospective comparative study. Ann Surg Oncol 46. Pompili M, Riccardi L, Covino M, et al. Contrast-
X, Lu DS. Microwave liver ablation: influence of 2011; 18:413420 enhanced gray-scale harmonic ultrasound in the
hepatic vein size on heat-sink effect in a porcine 33. Kulik LM, Carr BI, Mulcahy MF, et al. Safety efficacy assessment of ablation treatments for hepa-
model. J Vasc Interv Radiol 2008; 19:10871092 and efficacy of 90Y radiotherapy for hepatocel- tocellular carcinoma. Liver Int 2005; 25:954961
20. Shafiee H, Garcia PA, Davalos RV. A prelimi- lular carcinoma with and without portal vein 47. Baron RL, Oliver JH 3rd, Dodd GD 3rd, Nalesnik
nary study to delineate irreversible electropora- thrombosis. Hepatology 2008; 47:7181 M, Holbert BL, Carr B. Hepatocellular carcinoma:
tion from thermal damage using the Arrhenius 34. Avila MA, Berasain C, Sangro B, Prieto J. New evaluation with biphasic, contrast-enhanced, heli-
equation. J Biomech Eng 2009; 131:074509 therapies for hepatocellular carcinoma. Onco- cal CT. Radiology 1996; 199:505511
21. Llovet JM, Bruix J. Systematic review of ran- gene 2006; 25:38663884 48. Choi BI, Lee HJ, Han JK, Choi DS, Seo JB, Han
domized trials for unresectable hepatocellular 35. Llovet JM, Bruix J. Molecular targeted therapies MC. Detection of hypervascular nodular hepato-
carcinoma: chemoembolization improves sur- in hepatocellular carcinoma. Hepatology 2008; cellular carcinomas: value of triphasic helical
vival. Hepatology 2003; 37:429442 48:13121327 CT compared with iodized-oil CT. AJR 1997;
22. Llovet JM, Real MI, Montana X, et al. Arterial 36. Becker G, Allgaier HP, Olschewski M, Zahringer 168:219224
embolisation or chemoembolisation versus symp- A, Blum HE. Long-acting octreotide versus pla- 49. Iannaccone R, Laghi A, Catalano C, et al. Hepato-
tomatic treatment in patients with unresectable cebo for treatment of advanced HCC: a random- cellular carcinoma: role of unenhanced and delayed
hepatocellular carcinoma: a randomised con- ized controlled double-blind study. Hepatology phase multi-detector row helical CT in patients
trolled trial. Lancet 2002; 359:17341739 2007; 45:915 with cirrhosis. Radiology 2005; 234:460467
23. Lo CM, Ngan H, Tso WK, et al. Randomized 37. Chow PK, Tai BC, Tan CK, et al. High-dose 50. Wald C, Russo MW, Heimbach JK, Hussain HK,
controlled trial of transarterial Lipiodol chemo- tamoxifen in the treatment of inoperable hepato- Pomfret EA, Bruix J. New OPTN/UNOS policy
embolization for unresectable hepatocellular cellular carcinoma: a multicenter randomized con- for liver transplant allocation: standardization of
carcinoma. Hepatology 2002; 35:11641171 trolled trial. Hepatology 2002; 36:12211226 liver imaging, diagnosis, classification, and re-
24. Lammer J, Malagari K, Vogl T, et al.; Precision 38. Cheng AL, Kang YK, Chen Z, et al. Efficacy and porting of hepatocellular carcinoma. Radiology
V Investigators. Prospective randomized study safety of sorafenib in patients in the Asia-Pacific 2013; 266:376382
of doxorubicin-eluting-bead embolization in the region with advanced hepatocellular carcinoma: a 51. Silva AC, Morse BG, Hara AK, Paden RG, Hon-
treatment of hepatocellular carcinoma: results of phase III randomised, double-blind, placebo-con- go N, Pavlicek W. Dual-energy (spectral) CT: ap-
the PRECISION V study. Cardiovasc Intervent trolled trial. Lancet Oncol 2009; 10:2534 plications in abdominal imaging. RadioGraphics
Radiol 2010; 33:4152 39. Abou-Alfa GK, Johnson P, Knox JJ, et al. Doxo- 2011; 31:10311046; discussion, 10471050
25. Varela M, Real MI, Burrel M, et al. Chemoem- rubicin plus sorafenib vs doxorubicin alone in pa- 52. Zhang LJ, Peng J, Wu SY, et al. Liver virtual
bolization of hepatocellular carcinoma with tients with advanced hepatocellular carcinoma: a non-enhanced CT with dual-source, dual-energy

AJR:201, July 2013 93


Yaghmai et al.

CT: a preliminary study. Eur Radiol 2010; A):S28S36 sponse criteria best help predict survival of pa-
20:22572264 65. Khan MA, Combs CS, Brunt EM, et al. Positron tients with hepatocellular carcinoma following
53. Altenbernd J, Heusner TA, Ringelstein A, Ladd emission tomography scanning in the evaluation chemoembolization? A validation study of old
SC, Forsting M, Antoch G. Dual-energy-CT of of hepatocellular carcinoma. J Hepatol 2000; and new models. Radiology 2012; 262:708718
hypervascular liver lesions in patients with 32:792797 78. Kim BK, Kim KA, Park JY, et al. Prospective com-
HCC: investigation of image quality and sensi- 66. Talbot JN, Gutman F, Fartoux L, et al. PET/CT parison of prognostic values of modified Response
tivity. Eur Radiol 2011; 21:738743 in patients with hepatocellular carcinoma using Evaluation Criteria in Solid Tumours with Europe-
Downloaded from www.ajronline.org by 36.76.228.89 on 10/09/17 from IP address 36.76.228.89. Copyright ARRS. For personal use only; all rights reserved

54. Lee SH, Lee JM, Kim KW, et al. Dual-energy [(18)F]fluorocholine: preliminary comparison an Association for the Study of the Liver criteria in
computed tomography to assess tumor response with [(18)F]FDG PET/CT. Eur J Nucl Med Mol hepatocellular carcinoma following chemoemboli-
to hepatic radiofrequency ablation: potential di- Imaging 2006; 33:12851289 sation. Eur J Cancer 2013; 49:826834
agnostic value of virtual noncontrast images and 67. [No authors listed]. World Health Organization 79. Shim JH, Lee HC, Won HJ, et al. Maximum number
iodine maps. Invest Radiol 2011; 46:7784 (WHO) handbook for reporting results of cancer of target lesions required to measure responses to
55. Earls JP, Rofsky NM, DeCorato DR, Krinsky treatment. Geneva, Switzerland: WHO, 1979: transarterial chemoembolization using the enhance-
GA, Weinreb JC. Hepatic arterial-phase dynam- WHO offset publication no. 48 ment criteria in patients with intrahepatic hepatocel-
ic gadolinium-enhanced MR imaging: optimi- 68. Therasse P, Arbuck SG, Eisenhauer EA, et al. lular carcinoma. J Hepatol 2012; 56:406411
zation with a test examination and a power injec- New guidelines to evaluate the response to treat- 80. Gillmore R, Stuart S, Kirkwood A, et al. EASL
tor. Radiology 1997; 202:268273 ment in solid tumors: European Organization for and mRECIST responses are independent prog-
56. Cruite I, Schroeder M, Merkle EM, Sirlin CB. Research and Treatment of Cancer, National nostic factors for survival in hepatocellular can-
Gadoxetate disodiumenhanced MRI of the liver. Cancer Institute of the United States, National cer patients treated with transarterial emboliza-
Part 2. Protocol optimization and lesion appear- Cancer Institute of Canada. J Natl Cancer Inst tion. J Hepatol 2011; 55:13091316
ance in the cirrhotic liver. AJR 2010; 195:2941 2000; 92:205216 81. Kawaoka T, Aikata H, Murakami E, et al. Evalu-
57. Ringe KI, Husarik DB, Sirlin CB, Merkle EM. Gad- 69. Eisenhauer EA, Therasse P, Bogaerts J, et al. ation of the mRECIST and alpha-fetoprotein ratio
oxetate disodiumenhanced MRI of the liver. Part 1. New Response Evaluation Criteria in Solid Tu- for stratification of the prognosis of advanced-
Protocol optimization and lesion appearance in the mours: revised RECIST guideline (version 1.1). hepatocellular-carcinoma patients treated with
noncirrhotic liver. AJR 2010; 195:1328 Eur J Cancer 2009; 45:228247 sorafenib. Oncology 2012; 83:192200
58. Kamel IR, Bluemke DA, Eng J, et al. The role of 70. Suzuki C, Jacobsson H, Hatschek T, et al. Radio- 82. Kim YS, Rhim H, Lim HK. Imaging after radio-
functional MR imaging in the assessment of tu- logic measurements of tumor response to treat- frequency ablation of hepatic tumors. Semin Ul-
mor response after chemoembolization in pa- ment: practical approaches and limitations. Ra- trasound CT MR 2009; 30:4966
tients with hepatocellular carcinoma. J Vasc In- dioGraphics 2008; 28:329344 83. Memon K, Kulik L, Lewandowski RJ, et al. Ra-
terv Radiol 2006; 17:505512 71. Llovet JM, Di Bisceglie AM, Bruix J, et al. Design diographic response to locoregional therapy in
59. Kloeckner R, Otto G, Biesterfeld S, Oberholzer and endpoints of clinical trials in hepatocellular hepatocellular carcinoma predicts patient survival
K, Dueber C, Pitton MB. MDCT versus MRI as- carcinoma. J Natl Cancer Inst 2008; 100:698711 times. Gastroenterology 2011; 141:526535
sessment of tumor response after transarterial 72. Bruix J, Sherman M, Llovet JM, et al. Clinical 84. Virmani S, Ryu RK, Sato KT, et al. Effect of C-
chemoembolization for the treatment of hepato- management of hepatocellular carcinoma: con- arm angiographic CT on transcatheter arterial
cellular carcinoma. Cardiovasc Intervent Radiol clusions of the Barcelona-2000 EASL confer- chemoembolization of liver tumors. J Vasc In-
2010; 33:532540 enceEuropean Association for the Study of terv Radiol 2007; 18:13051309
60. Kierans AS, Elazzazi M, Braga L, et al. Ther- the Liver. J Hepatol 2001; 35:421430 85. Lewandowski RJ, Tepper J, Wang D, et al. MR
moablative treatments for malignant liver le- 73. Lencioni R, Llovet JM. Modified RECIST imaging perfusion mismatch: a technique to
sions: 10-year experience of MRI appearances (mRECIST) assessment for hepatocellular car- verify successful targeting of liver tumors dur-
of treatment response. AJR 2010; 194:523529 cinoma. Semin Liver Dis 2010; 30:5260 ing transcatheter arterial chemoembolization. J
61. Kim S, Mannelli L, Hajdu CH, et al. Hepatocel- 74. European Association for the Study of the Liver; Vasc Interv Radiol 2008; 19:698705
lular carcinoma: assessment of response to trans- European Organisation for Research and Treat- 86. Nishimine K, Uchida H, Matsuo N, et al. Seg-
arterial chemoembolization with image subtrac- ment of Cancer. EASL-EORTC clinical practice mental transarterial chemoembolization with
tion. J Magn Reson Imaging 2010; 31:348355 guidelines: management of hepatocellular carci- Lipiodol mixed with anticancer drugs for nonre-
62. Watanabe H, Kanematsu M, Goshima S, et al. Is noma. J Hepatol 2012; 56:908943 sectable hepatocellular carcinoma: follow-up
gadoxetate disodiumenhanced MRI useful for 75. Edeline J, Boucher E, Rolland Y, et al. Comparison CT and therapeutic results. Cancer Chemother
detecting local recurrence of hepatocellular car- of tumor response by Response Evaluation Criteria Pharmacol 1994; 33(suppl):S60S68
cinoma after radiofrequency ablation therapy? in Solid Tumors (RECIST) and modified RECIST 87. Nakazawa T, Kokubu S, Shibuya A, et al. Radio-
AJR 2012; 198:589595 in patients treated with sorafenib for hepatocellular frequency ablation of hepatocellular carcinoma:
63. Yoon JH, Lee EJ, Cha SS, et al. Comparison of ga- carcinoma. Cancer 2012; 118:147156 correlation between local tumor progression af-
doxetic acidenhanced MR imaging versus four- 76. Prajapati HJ, Spivey JR, Hanish SI, et al. mRECIST ter ablation and ablative margin. AJR 2007;
phase multi-detector row computed tomography in and EASL responses at early time point by contrast- 188:480488
assessing tumor regression after radiofrequency enhanced dynamic MRI predict survival in patients 88. Park MH, Rhim H, Kim YS, Choi D, Lim HK, Lee
ablation in subjects with hepatocellular carcinomas. with unresectable hepatocellular carcinoma (HCC) WJ. Spectrum of CT findings after radiofrequency
J Vasc Interv Radiol 2010; 21:348356 treated by doxorubicin drug-eluting beads transarte- ablation of hepatic tumors. RadioGraphics 2008;
64. Schima W, Ba-Ssalamah A, Kurtaran A, Schindl rial chemoembolization (DEB TACE). Ann Oncol 28:379390; discussion, 390392
M, Gruenberger T. Post-treatment imaging of 2012 Dec 5 [Epub ahead of print] 89. Kim SK, Lim HK, Kim YH, et al. Hepatocellular
liver tumours. Cancer Imaging 2007; 7(spec no 77. Shim JH, Lee HC, Kim SO, et al. Which re- carcinoma treated with radio-frequency ablation:

94 AJR:201, July 2013


Imaging Assessment of HCC Response to Therapy

spectrum of imaging findings. RadioGraphics BMC Cancer 2009; 9:208 17:11951200


2003; 23:107121 103. Faivre S, Zappa M, Vilgrain V, et al. Changes in 116. Kamel IR, Reyes DK, Liapi E, Bluemke DA, Ge-
90. Chung WS, Lee KH, Park MS, et al. Enhance- tumor density in patients with advanced hepato- schwind JF. Functional MR imaging assessment
ment patterns of hepatocellular carcinoma after cellular carcinoma treated with sunitinib. Clin of tumor response after 90Y microsphere treat-
transarterial chemoembolization using drug-elut- Cancer Res 2011; 17:45044512 ment in patients with unresectable hepatocellular
ing beads on arterial phase CT images: a pilot 104. Tran LN, Brown MS, Goldin JG, et al. Compari- carcinoma. J Vasc Interv Radiol 2007; 18:4956
retrospective study. AJR 2012; 199:349359 son of treatment response classifications be- 117. Kamel IR, Liapi E, Reyes DK, Zahurak M,
Downloaded from www.ajronline.org by 36.76.228.89 on 10/09/17 from IP address 36.76.228.89. Copyright ARRS. For personal use only; all rights reserved

91. Takayasu K, Arii S, Matsuo N, et al. Compari- tween unidimensional, bidimensional, and volu- Bluemke DA, Geschwind JF. Unresectable hepa-
son of CT findings with resected specimens after metric measurements of metastatic lung lesions tocellular carcinoma: serial early vascular and
chemoembolization with iodized oil for hepato- on chest computed tomography. Acad Radiol cellular changes after transarterial chemoembo-
cellular carcinoma. AJR 2000; 175:699704 2004; 11:13551360 lization as detected with MR imaging. Radiolo-
92. Kwan SW, Fidelman N, Ma E, Kerlan RK Jr, Yao 105. Galizia MS, Tore HG, Chalian H, Yaghmai V. gy 2009; 250:466473
FY. Imaging predictors of the response to trans- Evaluation of hepatocellular carcinoma size us- 118. Mannelli L, Kim S, Hajdu CH, Babb JS, Clark
arterial chemoembolization in patients with he- ing two-dimensional and volumetric analysis: TW, Taouli B. Assessment of tumor necrosis of
patocellular carcinoma: a radiological-patholog- effect on liver transplantation eligibility. Acad hepatocellular carcinoma after chemoemboliza-
ical correlation. Liver Transpl 2012; 18:727736 Radiol 2011; 18:15551560 tion: diffusion-weighted and contrast-enhanced
93. Kim HC, Chung JW, Lee W, Jae HJ, Park JH. 106. Yaghmai V, Miller FH, Rezai P, Benson AB 3rd, MRI with histopathologic correlation of the ex-
Recognizing extrahepatic collateral vessels that Salem R. Response to treatment series. Part 2. planted liver. AJR 2009; 193:10441052
supply hepatocellular carcinoma to avoid compli- Tumor response assessment: using new and con- 119. Parikh T, Drew SJ, Lee VS, et al. Focal liver le-
cations of transcatheter arterial chemoemboliza- ventional criteria. AJR 2011; 197:1827 sion detection and characterization with diffu-
tion. RadioGraphics 2005; 25(suppl 1):S25S39 107. Chalian H, Tochetto SM, Tore HG, Rezai P, sion-weighted MR imaging: comparison with
94. Okabe N, Morimoto M, Kondo M, et al. Efficacy Yaghmai V. Hepatic tumors: region-of-interest standard breath-hold T2-weighted imaging. Ra-
of chemoembolization for recurrent HCC after versus volumetric analysis for quantification of diology 2008; 246:812822
curative ablation therapy. Hepatogastroenterol- attenuation at CT. Radiology 2012; 262:853861 120. Goshima S, Kanematsu M, Kondo H, et al. Eval-
ogy 2012; 29:60 108. Alonzi R, Hoskin P. Functional imaging in clini- uating local hepatocellular carcinoma recurrence
95. Khankan AA, Murakami T, Onishi H, et al. He- cal oncology: magnetic resonance imaging and post-transcatheter arterial chemoembolization: is
patocellular carcinoma treated with radio fre- computerised tomographybased techniques. diffusion-weighted MRI reliable as an indicator?
quency ablation: an early evaluation with mag- Clin Oncol (R Coll Radiol) 2006; 18:555570 J Magn Reson Imaging 2008; 27:834839
netic resonance imaging. J Magn Reson Imaging 109. Piana G, Trinquart L, Meskine N, Barrau V, 121. Yuan Z, Ye XD, Dong S, et al. Role of magnetic
2008; 27:546551 Beers BV, Vilgrain V. New MR imaging criteria resonance diffusion-weighted imaging in evaluat-
96. Dromain C, de Baere T, Elias D, et al. Hepatic with a diffusion-weighted sequence for the diag- ing response after chemoembolization of hepato-
tumors treated with percutaneous radio-frequen- nosis of hepatocellular carcinoma in chronic cellular carcinoma. Eur J Radiol 2010; 75:e9e14
cy ablation: CT and MR imaging follow-up. Ra- liver diseases. J Hepatol 2011; 55:126132 122. Mannelli L, Kim S, Hajdu CH, Babb JS, Taouli
diology 2002; 223:255262 110. Park MS, Kim S, Patel J, et al. Hepatocellular B. Serial diffusion-weighted MRI in patients
97. Lim HK, Choi D, Lee WJ, et al. Hepatocellular car- carcinoma: detection with diffusion-weighted with hepatocellular carcinoma: prediction and
cinoma treated with percutaneous radio-frequency versus contrast-enhanced magnetic resonance assessment of response to transarterial chemo-
ablation: evaluation with follow-up multiphase he- imaging in pretransplant patients. Hepatology embolizationpreliminary experience. Eur J
lical CT. Radiology 2001; 221:447454 2012; 56:140148 Radiol 2012 Dec 12 [Epub ahead of print]
98. Riaz A, Kulik L, Lewandowski RJ, et al. Radiologic- 111. Koh DM, Collins DJ. Diffusion-weighted MRI 123. Schraml C, Schwenzer NF, Clasen S, et al. Navi-
pathologic correlation of hepatocellular carcinoma in the body: applications and challenges in on- gator respiratory-triggered diffusion-weighted
treated with internal radiation using yttrium-90 cology. AJR 2007; 188:16221635 imaging in the follow-up after hepatic radiofre-
microspheres. Hepatology 2009; 49:11851193 112. Hamstra DA, Rehemtulla A, Ross BD. Diffusion quency ablation: initial results. J Magn Reson
99. Ibrahim SM, Nikolaidis P, Miller FH, et al. Ra- magnetic resonance imaging: a biomarker for Imaging 2009; 29:13081316
diologic findings following Y90 radioemboliza- treatment response in oncology. J Clin Oncol 124. Kim SY, Lee SS, Byun JH, et al. Malignant he-
tion for primary liver malignancies. Abdom Im- 2007; 25:41044109 patic tumors: short-term reproducibility of ap-
aging 2009; 34:566581 113. Kamel IR, Bluemke DA, Ramsey D, et al. Role parent diffusion coefficients with breath-hold
100. Keppke AL, Salem R, Reddy D, et al. Imaging of of diffusion-weighted imaging in estimating tu- and respiratory-triggered diffusion-weighted
hepatocellular carcinoma after treatment with yt- mor necrosis after chemoembolization of hepa- MR imaging. Radiology 2010; 255:815823
trium-90 microspheres. AJR 2007; 188:768775 tocellular carcinoma. AJR 2003; 181:708710 125. Kim SY, Lee SS, Park B, et al. Reproducibility
101. Kim MJ, Choi JI, Lee JS, Park JW. Computed to- 114. Chen CY, Li CW, Kuo YT, et al. Early response of measurement of apparent diffusion coeffi-
mography findings of sorafenib-treated hepatic of hepatocellular carcinoma to transcatheter ar- cients of malignant hepatic tumors: effect of
tumors in patients with advanced hepatocellular terial chemoembolization: choline levels and DWI techniques and calculation methods. J
carcinoma. J Gastroenterol Hepatol 2011; 26: MR diffusion constantsinitial experience. Ra- Magn Reson Imaging 2012; 36:11311138
12011206 diology 2006; 239:448456 126. Padhani AR. Dynamic contrast-enhanced MRI
102. Horger M, Lauer UM, Schraml C, et al. Early 115. Deng J, Miller FH, Rhee TK, et al. Diffusion- studies in human tumours. Br J Radiol 1999;
MRI response monitoring of patients with ad- weighted MR imaging for determination of he- 72:427431
vanced hepatocellular carcinoma under treat- patocellular carcinoma response to yttrium-90 127. Padhani AR. Dynamic contrast-enhanced MRI in
ment with the multikinase inhibitor sorafenib. radioembolization. J Vasc Interv Radiol 2006; clinical oncology: current status and future direc-

AJR:201, July 2013 95


Yaghmai et al.

tions. J Magn Reson Imaging 2002; 16:407422 Sahani DV. Monitoring response to antiangio- tent triple angiokinase inhibitor. Eur Radiol
128. Parker GJ, Suckling J, Tanner SF, et al. Probing genic treatment and predicting outcomes in ad- 2008; 18:14141421
tumor microvascularity by measurement, analy- vanced hepatocellular carcinoma using image bio- 139. Abdullah SS, Pialat JB, Wiart M, et al. Charac-
sis and display of contrast agent uptake kinetics. markers, CT perfusion, tumor density, and tumor terization of hepatocellular carcinoma and colo-
J Magn Reson Imaging 1997; 7:564574 size (RECIST). Invest Radiol 2012; 47:1117 rectal liver metastasis by means of perfusion
129. Petralia G, Summers P, Viotti S, Montefrances- 135. Morgan B, Thomas AL, Drevs J, et al. Dynamic MRI. J Magn Reson Imaging 2008; 28:390395
co R, Raimondi S, Bellomi M. Quantification of contrast-enhanced magnetic resonance imaging 140. Jarnagin WR, Schwartz LH, Gultekin DH, et al.
Downloaded from www.ajronline.org by 36.76.228.89 on 10/09/17 from IP address 36.76.228.89. Copyright ARRS. For personal use only; all rights reserved

variability in breath-hold perfusion CT of hepa- as a biomarker for the pharmacological response Regional chemotherapy for unresectable prima-
tocellular carcinoma: a step toward clinical use. of PTK787/ZK 222584, an inhibitor of the vascu- ry liver cancer: results of a phase II clinical trial
Radiology 2012; 265:448456 lar endothelial growth factor receptor tyrosine and assessment of DCE-MRI as a biomarker of
130. Stewart EE, Sun H, Chen X, et al. Effect of an kinases, in patients with advanced colorectal can- survival. Ann Oncol 2009; 20:15891595
angiogenesis inhibitor on hepatic tumor perfu- cer and liver metastases: results from two phase I 141. Yopp AC, Schwartz LH, Kemeny N, et al. Antian-
sion and the implications for adjuvant cytotoxic studies. J Clin Oncol 2003; 21:39553964 giogenic therapy for primary liver cancer: correla-
therapy. Radiology 2012; 264:6877 136. Wang J, Chen LT, Tsang YM, Liu TW, Shih TT. tion of changes in dynamic contrast-enhanced
131. Sahani DV, Holalkere NS, Mueller PR, Zhu AX. Dynamic contrast-enhanced MRI analysis of magnetic resonance imaging with tissue hypoxia
Advanced hepatocellular carcinoma: CT perfu- perfusion changes in advanced hepatocellular markers and clinical response. Ann Surg Oncol
sion of liver and tumor tissueinitial experi- carcinoma treated with an antiangiogenic agent: 2011; 18:21922199
ence. Radiology 2007; 243:736743 a preliminary study. AJR 2004; 183:713719 142. Wang D, Gaba RC, Jin B, et al. Intraprocedural
132. Ippolito D, Sironi S, Pozzi M, et al. Perfusion 137. Totman JJ, OGorman RL, Kane PA, Karani JB. transcatheter intra-arterial perfusion MRI as a
computed tomographic assessment of early he- Comparison of the hepatic perfusion index mea- predictor of tumor response to chemoemboliza-
patocellular carcinoma in cirrhotic liver disease: sured with gadolinium-enhanced volumetric tion for hepatocellular carcinoma. Acad Radiol
initial observations. J Comput Assist Tomogr MRI in controls and in patients with colorectal 2011; 18:828836
2008; 32:855858 cancer. Br J Radiol 2005; 78:105109 143. Taouli B, Johnson RS, Hajdu C, et al. Hepatocellular
133. Ippolito D, Sironi S, Pozzi M, et al. Perfusion CT 138. Miyazaki K, Collins DJ, Walker-Samuel S, et al. carcinoma: perfusion quantification with 3D perfu-
in cirrhotic patients with early stage hepatocel- Quantitative mapping of hepatic perfusion index sion-weighted MR imaging. AJR 2013 (in press)
lular carcinoma: assessment of tumor-related using MR imaging: a potential reproducible tool 144. Thng CH, Koh TS, Collins DJ, Koh DM. Perfu-
vascularization. Eur J Radiol 2010; 73:148152 for assessing tumour response to treatment with sion magnetic resonance imaging of the liver.
134. Jiang T, Kambadakone A, Kulkarni NM, Zhu AX, the antiangiogenic compound BIBF 1120, a po- World J Gastroenterol 2010; 16:15981609

96 AJR:201, July 2013


This article has been cited by:

1. H.L. Lewis, M.A. Ghasabeh, P. Khoshpouri, I.R. Kamel, T.M. Pawlik. 2017. Functional hepatic imaging as a biomarker of
primary and secondary tumor response to loco-regional therapies. Surgical Oncology 26:4, 411-422. [Crossref]
2. R.H. Jones, A.J. Taylor, N. Rostambeigi, B. Spilseth. 2017. Small hepatocellular carcinomas displayed as a ring enhancing mass
on arterial phase MRI in the chronically diseased liver. Clinical Radiology 72:11, 995.e1-995.e9. [Crossref]
3. Jakob Weiss, Hansjoerg Rempp, Stephan Clasen, Mike Notohamiprodjo, David-Emanuel Keler, Philippe L. Pereira, Gunnar
Downloaded from www.ajronline.org by 36.76.228.89 on 10/09/17 from IP address 36.76.228.89. Copyright ARRS. For personal use only; all rights reserved

Blumenstock, Konstantin Nikolaou, Rdiger Hoffmann. 2017. Diagnostic accuracy of different magnetic resonance imaging
sequences for detecting local tumor progression after radiofrequency ablation of hepatic malignancies. European Journal of
Radiology 94, 85-92. [Crossref]
4. Ania Kielar, Kathryn J. Fowler, Sara Lewis, Vahid Yaghmai, Frank H. Miller, Hooman Yarmohammadi, Charles Kim, Victoria
Chernyak, Takeshi Yokoo, Jeffrey Meyer, Isabel Newton, Richard K. Do. 2017. Locoregional therapies for hepatocellular
carcinoma and the new LI-RADS treatment response algorithm. Abdominal Radiology 273. . [Crossref]
5. N. Patel, A.J. King, D.J. Breen. 2017. Imaging appearances at follow-up after image-guided solid-organ abdominal tumour
ablation. Clinical Radiology 72:8, 680-690. [Crossref]
6. Anne M. Covey, Shahid M. Hussain. 2017. Liver-Directed Therapy for Hepatocellular Carcinoma: An Overview of Techniques,
Outcomes, and Posttreatment Imaging Findings. American Journal of Roentgenology 209:1, 67-76. [Abstract] [Full Text] [PDF]
[PDF Plus]
7. Leigh Anne Dageforde, Kathryn J. Fowler, William C. Chapman. 2017. Liver transplantation for hepatocellular carcinoma.
Current Opinion in Organ Transplantation 22:2, 128-134. [Crossref]
8. Xiao-Dan Ye, Zuguo Yuan, Jian Zhang, Zheng Yuan. 2017. Radiological biomarkers for assessing response to locoregional
therapies in hepatocellular carcinoma: From morphological to functional imaging (Review). Oncology Reports . [Crossref]
9. Atilla Arslanoglu, Adeel R. Seyal, Faezeh Sodagari, Azize Sahin, Frank H. Miller, Riad Salem, Vahid Yaghmai. 2016. Current
Guidelines for the Diagnosis and Management of Hepatocellular Carcinoma: A Comparative Review. American Journal of
Roentgenology 207:5, W88-W98. [Abstract] [Full Text] [PDF] [PDF Plus]
10. Hazim I. Tantawy, Faten Fawzy Mohamed. 2016. Diagnostic value of apparent diffusion coefficient (ADC) in evaluating
hepatocellular carcinomas post trans-catheter arterial chemoembolization and radiofrequency ablation. The Egyptian Journal of
Radiology and Nuclear Medicine 47:3, 699-706. [Crossref]
11. Joseph Ralph Kallini, Frank H. Miller, Ahmed Gabr, Riad Salem, Robert J. Lewandowski. 2016. Hepatic imaging following
intra-arterial embolotherapy. Abdominal Radiology 41:4, 600-616. [Crossref]
12. Danny Ngo, Jemianne Bautista Jia, Christopher S. Green, Anjalie T. Gulati, Chandana Lall. 2015. Cancer therapy related
complications in the liver, pancreas, and biliary system: an imaging perspective. Insights into Imaging 6:6, 665-677. [Crossref]
13. Johannes Ludwig, Juan Camacho, Nima Kokabi, Minzhi Xing, Hyun Kim. 2015. The Role of Diffusion-Weighted Imaging
(DWI) in Locoregional Therapy Outcome Prediction and Response Assessment for Hepatocellular Carcinoma (HCC): The
New Era of Functional Imaging Biomarkers. Diagnostics 5:4, 546-563. [Crossref]
14. Surabhi Bajpai, Avinash Kambadakone, Alexander R. Guimaraes, Ronald S. Arellano, Debra A. Gervais, Dushyant Sahani. 2015.
Image-guided Treatment in the Hepatobiliary System: Role of Imaging in Treatment Planning and Posttreatment Evaluation.
RadioGraphics 35:5, 1393-1418. [Crossref]
15. Jeet Minocha, Robert J. Lewandowski. 2015. Assessing Imaging Response to Therapy. Radiologic Clinics of North America
53:5, 1077-1088. [Crossref]
16. Tommaso Vincenzo Bartolotta, Adele Taibbi, Domenica Matranga, Massimo Midiri, Roberto Lagalla. 2015. 3D versus 2D
contrast-enhanced sonography in the evaluation of therapeutic response of hepatocellular carcinoma after locoregional therapies:
preliminary findings. La radiologia medica 120:8, 695-704. [Crossref]
17. Juliane Schelhorn, Jan Best, Marcus P. Reinboldt, Guido Gerken, Marcus Ruhlmann, Thomas C. Lauenstein, Gerald Antoch,
Sonja Kinner. 2015. Therapy Response Assessment after Radioembolization of Patients with Hepatocellular Carcinoma
Comparison of MR Imaging with Gadolinium Ethoxybenzyl Diethylenetriamine Penta-Acetic Acid and Gadobutrol. Journal of
Vascular and Interventional Radiology 26:7, 972-979. [Crossref]
18. Nima Kokabi, Juan C. Camacho, Minzhi Xing, Faramarz Edalat, Pardeep K. Mittal, Hyun S. Kim. 2015. Immediate post-
doxorubicin drug-eluting beads chemoembolization Mr Apparent diffusion coefficient quantification predicts response in
unresectable hepatocellular carcinoma: A pilot study. Journal of Magnetic Resonance Imaging n/a-n/a. [Crossref]
19. Daniel Marin, Salvatore Cappabianca, Nicola Serra, Assunta Sica, Francesco Lassandro, Roberto DAngelo, Michelearcangelo La
Porta, Francesco Fiore, Francesco Somma. 2015. CT Appearance of Hepatocellular Carcinoma after Locoregional Treatments:
A Comprehensive Review. Gastroenterology Research and Practice 2015, 1-10. [Crossref]
20. Nima Kokabi, Juan C. Camacho, Minzhi Xing, Deqiang Qiu, Hiroumi Kitajima, Pardeep K. Mittal, Hyun S. Kim. 2014.
Apparent diffusion coefficient quantification as an early imaging biomarker of response and predictor of survival following
yttrium-90 radioembolization for unresectable infiltrative hepatocellular carcinoma with portal vein thrombosis. Abdominal
Imaging 39:5, 969-978. [Crossref]
Downloaded from www.ajronline.org by 36.76.228.89 on 10/09/17 from IP address 36.76.228.89. Copyright ARRS. For personal use only; all rights reserved

21. Sara Lewis, Hadrien Dyvorne, Yong Cui, Bachir Taouli. 2014. Diffusion-Weighted Imaging of the Liver. Magnetic Resonance
Imaging Clinics of North America 22:3, 373-395. [Crossref]

Anda mungkin juga menyukai