Review Article

Modern multimodality diagnosis of portal vein infiltration in hepatocellular carcinoma and expected changes during current therapies

Acta Radiologica 2015, Vol. 56(11) 1283–1292 ! The Foundation Acta Radiologica 2014 Reprints and permissions: sagepub.co.uk/journalsPermissions.nav DOI: 10.1177/0284185114556305 acr.sagepub.com

Sascha Kaufmann, Maximilian Schulze, Daniel Spira and Marius Horger

Abstract Diagnosis of malignant portal vein infiltration and especially differentiation from bland thrombosis is challenging for most imaging techniques. The use of contrast-enhanced real-time modern imaging modalities like contrast-enhanced ultrasound (CEUS), volume perfusion CT (VPCT), or dynamic contrast-enhanced MRI (DCE-MRI) with dedicated postprocessing tools provides additional support in difficult cases and is therefore recommended. The purpose of this review is to present a practical overview of strengths and limitations of modern imaging techniques in the diagnosis of malignant infiltration of the portal vein by hepatocellular carcinoma and to describe subsequent changes during therapy.

Keywords Liver, portal vein thrombosis, computed tomography (CT), magnetic resonance imaging (MRI), ultrasound Date received: 2 December 2013; accepted: 22 September 2014

Introduction Hepatocellular carcinoma (HCC) represents the fifth most common cancer worldwide and the second most common cause of cancer-related mortality making out 90% of primary malignant neoplasm of the liver (1–3). Tumor invasion of the portal vein as well as benign portal vein thrombosis (PVT) are both associated with HCC occurring in the setting of liver cirrhosis and portal hypertension with a prevalence of 44% and 42%, respectively (4,5). Moreover, these complications do often co-exist. Differentiation between malignant infiltration of the portal vein and PVT has great impact on prognosis of HCC according to the Barcelona Clinic Liver Cancer (BCLC) staging which is significantly poorer (advanced stage-C) in patients with malignant PVT with a mean survival time of 2.7–4.0 months (6,7). Venous invasion is associated with higher grade and larger tumors and represents an independent predictor of survival (6). Moreover, malignant infiltration of the portal vein in HCC is usually an exclusion criterion for aggressive treatments like

transarterial chemoembolization (TACE) and liver surgery or orthotopic liver transplantation due to high recurrence and complication rates (5). In this clinical setting, recent data showed that TACE can be safely performed in advanced HCC patients with vascular invasion improving overall survival (8,9). Even repeated TACE sessions seem to have no negative impact on the liver function (8). Positive results have also been reported for other local therapy techniques like radiofrequency ablation (RFA), transarterial radioembolization (TARE), hepatic artery infusional chemotherapy with 5-fluorouracil, TACE and portal vein and stent combined with 125I seed, Department of Radiology, Diagnostic and Interventional Radiology, Eberhard-Karls-University, Tu¨bingen, Germany Corresponding author: Sascha Kaufmann, MD, Department of Radiology, Diagnostic and Interventional Radiology, Eberhard-Karls-University, Hoppe-Seyler-Str. 3, 72076 Tu¨bingen, Germany. Email: [email protected]

Downloaded from acr.sagepub.com at BOSTON UNIV on October 18, 2015

1284

Acta Radiologica 56(11)

three-dimensional (3D) conformal radiotherapy combined with TACE, as well as surgical resection and systemic treatment with sorafenib combined with TACE (9–16). Two different pathways of malignant portal vein infiltration are recognized: tumor invasion into the portal vein by direct venous extension or by hematogenous seeding. In the latter, there is no apparent anatomic relation between the intraluminal portal vein mass and the primary hepatic tumor making diagnosis particularly challenging. Malignant infiltrates of the portal vein have their own arterial supply stemming from the vasa vasorum of the portal vein (17). The gold standard for characterization of a portal venous infiltration is histopathological examination. However, due to the invasive nature of biopsy and associated bleeding risk in liver cirrhosis, differentiation in clinical routine is usually based on imaging findings (18). Furthermore, imaging is crucial not only for diagnosis, but also for evaluation of response to therapy. According to Response Evaluation Criteria in Solid Tumors (RECIST) which are based on size measurements of tumor target lesions, even focal malignant infiltration of the portal vein is considered non-target. This is due to difficulties in determining its nature, and the poor measurability because of its amorphous configuration (19). New therapeutic agents exhibiting antiangiogenic properties often show little influence on tumor size, but rather affect other tumor parameters like vascularization or water diffusivity. Hence, the combined use of morphologic and functional imaging techniques becomes imperative. The most frequently used diagnostic imaging tools are ultrasound (US) with Doppler mode, contrast-enhanced ultrasound (CEUS), contrast-enhanced computed tomography (CECT), contrast-enhanced magnetic resonance imaging (MRI), and fluorodeoxyglucose positron emission tomography (FDG-PET).

Ultrasound, color Doppler, power Doppler, and contrast-enhanced ultrasound The echogenicity of a thrombus is a poor discriminator between malignant infiltration of the portal vein and bland PVT (Figs. 1–3). Thrombus echogenicity is increased in both disorders, comparable to primary hepatic tumors or adjacent parenchyma, but variable echogenicity can also be encountered (Figs. 1a, 2a, and 3a). With time, echogenicity decreases in bland PVT paralleling volume loss, contrary to malignant infiltration of the portal vein which is expected to farther expand and demonstrate neovascularization if treated inefficiently (20). Flow visualization of the portal vein using CD and PD may confirm lack of

patency with either missing flow signal or presence of pulsatile flow due to a-v shunts between hepatic artery (HA) and portal vein (VP), as well as collateralization. Collateralization usually recruits and dilates the preformed peribiliary venous plexus (Figs. 2b and 3b). Moreover, some HCC-induced infiltrates of the portal vein are highly vascularized and therefore simulate normal flow in the portal vein on CD or PD if no spectral analysis is performed. Similarly, post-thrombotic syndromes showing partial recanalization of the vessel lumen sometimes mimic either marked vascularization of intravascular tumor or normal vessel patency. In such cases, the use of pulsed Doppler with spectral analysis can disclose arterial flow characteristics of the tumor infiltrating the portal vein. With the advent of second generation US contrast agents, the clinical practice of HCC evaluation has been revolutionized and reports attest higher sensitivity and accuracy for the diagnosis of hepatic malignancies with CEUS compared to standardized CECT (Fig. 1c and d) (21). Early enhancement of a mass within the portal vein lumen on CEUS is an indicator of malignant infiltration (Fig. 2c and d) (20). In conformity with established criteria used for non-invasive characterization of HCC through other imaging techniques, the presence of contrast washout of the thrombus in the venous phase on CEUS is suggestive for malignant PVT (22). Fresh bland PVT shows no intraluminal flow on CEUS (Fig. 3b). In contrast to CT or MRI contrast agents, microbubbles used in CEUS remain in the intravascular space, thus displaying tumor vascularization rather than extravascular diffusion of the contrast agent. For CEUS a specificity of 100% and a sensitivity of 83% are reported for detection and discrimination of malignant infiltration of the portal vein. The lower sensitivity of CEUS due to limitations like patient’s body habitus and operator experience may limit its use in some cases. During antiangiogenic therapy, CEUS reveals decreasing vascularization and potential recanalization of the portal vein, paralleling changes in the primary tumor (Fig. 1e). If the tumor completely regresses, monophasic venous flow can be detected on CD reflecting partially recovered portal vein patency.

Computed tomography, volume perfusion CT, and PET-CT In unenhanced CT, malignant and fresh benign intravascular portal vein thrombosis produces a hypo- to isoattenuating filling defect expanding the vascular lumen (Figs. 4–6). Most HCCs are slightly hypoattenuating on precontrast images, except for rare spontaneous hemorrhage which may cause hyperattenuation. On CECT, enhancement of an intraluminal mass usually discriminates between malignant infiltration of the

Downloaded from acr.sagepub.com at BOSTON UNIV on October 18, 2015

Kaufmann et al.

1285

Fig. 1. A 61-year-old male patient with histologically proven HCC showing increased echogenicity of the portal vein lumen on Bmode liver US (arrows, a), suggesting benign or malignant infiltration of the portal vein. The lack of patency of the portal vein could be easily confirmed by power Doppler (PD). However, within the portal vein no intravascular flow was detected (arrow) but collateralization along the venous peribiliary plexus (color-coded flow) (b). Using CEUS, intravascular flow of microbubbles (arrows) could be demonstrated both, in the arterial (c) and capillary perfusion phase (d) of the liver confirming the diagnosis of malignant PVT. Also note intraparenchymal large HCC next to the portal vein, invading the vessel (left lower quadrant). The adjacent normal liver parenchyma showed comparable low vascularization at that time. At follow-up 12 weeks later, following systemic monotherapy with the VEGF-R inhibitor bevacizumab, intratumoral flow (arrow) significantly decreased and was still restricted to the thickened vessel wall infiltration and vicinity (e).

portal vein and bland PVT. Rarely, enhancement may be encountered even in bland PVT reflecting collateral channels in or along a blood clot or slow flow in an organized thrombus with intrinsic vascularization, challenging the radiologist (Figs. 4a, 5a and 6a) (23). Moreover, subtle changes in CT attenuation of malignant infiltration of the portal vein reflecting neovascularization can be missed by double-phase CT and differentiation from partial volume averaging due to enhancing adjacent liver parenchyma, or partially patent portal vein lumen, may hamper such measurements. Multi-phase CT with image subtraction might represent a solution for detection of neovascularization in malignant infiltration of the portal vein, similar to that measured in the primary tumor (HCC). Multiphase CECT using iodine contrast agent, holds a sensitivity of 86 % and a specificity of 100% for detecting HCC, comparable to that of CEUS (23,24). Current international guidelines define a typical HCC vascular pattern (assessed by non-invasive imaging techniques) as a homogeneous hyperenhancement (wash-in) in the arterial phase followed by washout in the venous or late

phase (25). Therefore, multi-phase CECT has gained great popularity on this diagnostic field. Nonetheless, some HCCs do not fulfil these criteria (wash-in/washout). Besides, detection of wash-in is strongly dependent on the chosen delay time, circulation time, viscosity of contrast media, etc. For this purpose, a multi-arterial (volume perfusion CT [VPCT]) protocol is more likely to detect these tumor characteristics. Furthermore, the main challenge in cirrhotic patients is differentiation between regenerative liver nodules and less vascularized HCCs (e.g. well-differentiated subtypes). Whereas regenerative hepatic nodules are supposed to be entirely supplied by the portal vein, HCCs become more and more arterialized with progressive dedifferentiation. VPCT as a novel technique measures temporal changes in tissue density after intravenous contrast injection using repeated CT scans of the volume being analyzed. Different kinetic modeling techniques are used by software providers. A one-compartment analysis of short-duration scans during the initial phase after contrast injection allows the calculation of perfusion (blood flow [BF]) and time to peak (TTP), whereas

Downloaded from acr.sagepub.com at BOSTON UNIV on October 18, 2015

1286

Acta Radiologica 56(11)

Fig. 2. A 76-year-old male patient with known HCC and segmental distension of portal vein lumen of unknown origin. On B-mode US image (a), papillary projections into the portal vein could be detected (arrows). Corresponding color Doppler (CD) demonstrated intravascular flow through the patent vessel lumen (arrowhead) but, no vascularization (arrow) inside the suspected mural nodules (b). After intravenous administration of US contrast agent, marked, early (arterial) accumulation of microbubbles is seen in the suspected area demonstrating tumor invasion of the portal vein (arrow) (c). Note similar contralateral tumor infiltration (arrowhead). Fifteen seconds later, intraluminal flow is seen (long slim arrow) through the patent portal vein in addition to the arterial flow in the mural tumor infiltrate (arrow and contralateral arrowhead) (d).

Fig. 3. A 68-year-old female patient with HCC and PVT of unknown origin. Marked luminal expansion and increased intraluminal echogenicity are seen on B-mode US (a); but they are both unspecific. With CEUS, lack of intraluminal flow (arrow) as well as incipient collateralization of the venous peribiliary plexus (arrowhead) are noticed (b). At follow-up, spontaneous shrinkage of the bland thrombus (arrow) was documented confirming the diagnosis of benign PVT (c).

Downloaded from acr.sagepub.com at BOSTON UNIV on October 18, 2015

Kaufmann et al.

1287

Fig. 4. A 67-year-old male patient with HCC and newly diagnosed malignant PVT. On contrast-enhanced arterial (a) and portal– venous (b) phase CT (CECT), a slightly hypoattenuating mass is seen occluding the portal vein (arrows). Notice the enhancing HCC in the liver parenchyma next to it (arrowhead). VPCT shows increased BF (arrow, c) and BV (arrow, d) and nicely delineates the primary tumor (arrowhead). On the portal–venous perfusion color-coded map (e), solely portal–venous vascularized liver parenchyma is displayed. Notice that the malignant infiltration of the portal vein is showing no perfusion. Following combined systemic therapy with the multitargeted thyrosine kinase inhibitor sorafenib and the epidermal growth factor receptor EGFR1 inhibitor erlotinib, marked hypoattenuation of the infiltrating tumor in the portal vein (arrows) is seen (f). Correspondingly, VPCT parameters showed decreased values at follow-up (not shown). Also note the prominent collateralization of the venous peribiliary plexus (black arrowhead). In the right liver lobe, a typical wedge resection area is seen after previous surgery.

Downloaded from acr.sagepub.com at BOSTON UNIV on October 18, 2015

1288

Acta Radiologica 56(11)

Fig. 5. A 78-year-old male patient with known HCC and newly occurred malignant PVT of the right portal vein branch (arrow) of unknown origin documented on arterial (a) and portal–venous postcontrast phase (b). Note high perfusion (arrows) of occluded portal vein on BF color-coded map (c), as well as some patchy infiltration of the liver parenchyma by diffuse HCC that was not obvious on CECT.

a two-compartment (Patlak) analysis quantifies exchanges between intra- and extravascular spaces, enabling calculation of blood volume ([BV], excluding stagnant blood) and permeability (PMB) of vessels. The latter is also known as transit constant ([Ktrans], defined as the sum of the flow within the microvasculature and capillary permeability) (26,27). Additionally, separate calculation of arterial and portal–venous liver perfusion (ALP and PVP) by appropriate placement of ROIs in the portal vein and the spleen is possible due to the simultaneous perfusion of both organs via hepatic and splenic arteries. This is analogous to former CT hepatic arteriography (CTHA) and CT arterioportography (CTAP) techniques, but can be performed in a non-invasive way. This separate illustration of hepatic arterial and portal–venous blood supply via VPCT is further increasing sensitivity and specificity disclosing exclusive arterialization of otherwise poorly vascularized tumors. VPCT has become an important biomarker for monitoring treatment response, especially to antiangiogenic drugs (28). Previous studies focused on the role of perfusion CT for discrimination between well-differentiated and moderately or poorly differentiated HCC (29). It was shown that perfusion CT techniques can quantify tumor perfusion parameters like BF, BV, or Ktrans (Figs. 4b, c, 5b, 6b, c, and 7a, b) (30,31). Of note, non-invasive diagnostic criteria for HCC are fully transferable to the diagnosis of malignant PVT. Hence, highly arterialized processes infiltrating the portal vein (high hepatic arterial perfusion index) lacking portal–venous perfusion are diagnostic for tumor infiltration. These characteristics allow better detection of additional inconspicuous hepatic HCC sites and improve monitoring response to systemic or local therapy. Here, declining perfusion values reflect either tumor necrosis or a temporary, drug-induced ‘switch off’ in tumor angiogenesis (Figs. 4d, 6c). Sophisticated motion correction algorithms enable accurate recognition of vessel invasion and

quantification of perfusion parameters. Therefore, this method should be used either in primary diagnosis of HCC when portal vein invasion is suspected or in equivocal cases (e.g. if CEUS and/or CECT have failed). The only disadvantage of this modality is radiation exposure, which is slightly higher for VPCT. Nevertheless, this aspect pales in comparison to the disease course and its mean survival time, especially in case of palliative treatment. In contrast to the other imaging techniques described showing morphological changes in tissues, 18F-FDG-PET is able to detect metabolic changes in tumors. Few data exist on the use of PET-CT when trying to differentiate malignant infiltration of the portal vein from benign PVT, but the current experiences with FDG-PET for HCC diagnosis argue against its routine application in this clinical setting (32–34).

Magnetic resonance imaging Malignant portal vein infiltration can exhibit variable signal intensities, but usually presents with hypo- to isointense signal on T1-weighted (T1W) images and slightly hyper- to isointense signal on T2-weighted (T2W) images (Fig. 8 a and b). In contrast, bland PVT often shows low signal intensity on T2W images due to its hemosiderin content. Li et al. reported comparable results of T2*W-weighted (T2*W) imaging as compared to postcontrast studies for characterization of benign PVT (35). However, differentiation of malignant from benign intravascular portal vein processes on the basis of MRI signal intensity alone remains difficult. Similar to CT, MRI diagnosis of HCC is mainly based on the evidence of contrast enhancement in tumor (36). On postcontrast studies of malignant infiltration of the portal vein, an early increase in signal intensity (arterialization) on fat-saturated T1W scans followed by progressive washout on later dynamic phases is typical (Fig. 8c). Beside gadopentat dimeglumin

Downloaded from acr.sagepub.com at BOSTON UNIV on October 18, 2015

Kaufmann et al.

1289

Fig. 6. A 68-year-old male patient with HCC and PVT of unknown origin. CECT shows slightly hypoattenuating thrombus of the left portal vein branch (arrows) in the arterial (a) and portal–venous (b) postcontrast phases. VPCT shows lack of perfusion on BF (c) and BV (d) color-coded maps (arrows) despite large HCC next to it in the liver segment 3. At follow-up (e), T1W MRI sequence illustrates typical methemoglobin-related hyperintense signal and slight shrinkage of the PVT despite adjacent HCC confirming the diagnosis of benign PVT.

(GD-DPTA), hepatocellular contrast agents (e.g. gadoxetate disodium) have gained in popularity for the diagnosis of HCC and related pathologies (e.g. malignant infiltration of the portal vein) due to a better tumor to tissue contrast (36). Their use enhances the sensitivity to non-invasively diagnose small hepatocellular carcinoma nodules in cirrhotic patients. Double hypointensity in the portal/venous and hepatobiliary phases has been proposed as a new MR pattern, highly suggestive of hypovascular hepatocellular carcinoma (37). By some authors the signal intensity increase in the dynamic postcontrast T1W images is still considered superior for extracellular contrast

media when compared to hepatocellular-specific agents. In equivocal cases, image subtraction of the contrast-enhanced dynamic series from the unenhanced scan has proved beneficial, as long as not limited by motion or breathing artifacts (Fig. 8d). Sandrasegaran et al. showed that the presence of at least two of the three following MRI findings had a sensitivity of 100% and a specificity of 90% for the diagnosis of malignant PVT: distance from tumor to PVT of less than 2 cm; HCC size of greater than 5 cm; and arterial enhancement of PVT (38). Similar to intrahepatic HCC, intravascular tumor infiltration may often but not always present restriction of water diffusivity with

Downloaded from acr.sagepub.com at BOSTON UNIV on October 18, 2015

1290

Acta Radiologica 56(11)

Fig. 7. A 60-year-old male patient with known HCC. Following previous surgery for focal HCC, PVT of unknown origin was newly diagnosed by CECT (a). Note marked venous collateralization on both sides of the portal vein lumen. Perfusion of the process infiltrating the portal vein was presumed on the basis of CECT. However, VPCT demonstrates arterial flow (arrow) in the portal vein lumen and the relapsing HCC in the right liver lobe adjacent to the previously resected tumor, representing malignant PVT (b). Also note dilatation of the peribiliary venous plexus within the scope of collateralization and tumor-induced occlusion of the portal vein.

Fig. 8. A 70-year-old male patient with known HCC and malignant PVT. On T1W (a) and T2W images (b) infiltration of the portal vein is demonstrated (arrows). However the cause for the vessel occlusion remains unclear. After intravenous contrast agent administration, slight enhancement (arrow) comparable to that of the adjacent liver parenchyma is seen (c). Corresponding diffusionweighted images (d) and ADC maps (e) reveal heterogeneous signal, but no evident restriction of water diffusivity. Six weeks later, following oral administration of a multi-targeted tyrosine kinase inhibitor, focally increased signal intensity (black arrows) on nonenhanced T1W image (f) as well as decreased enhancement on postcontrast fat-saturated T1W image (g) is seen. Six months later, postcontrast fat-saturated T1W image demonstrates revascularization of the malignant process infiltrating the portal vein as an expression of tumor re-growth (arrows, h).

correspondingly low apparent diffusion coefficient (ADC) which may aid in the discrimination of benign from malignant PVT (18) (Fig. 8e). Successful therapy increases water diffusivity and results in a higher ADC value, which reflects derangement of macromolecular

tumor architecture (e.g. due to necrosis). Interestingly, Maksimovic et al. demonstrated that early changes in ADC values in HCC patients treated with sorafenib show a contrary trend (39). Hence, acute PVT may also exhibit a decrease in ADC caused by shrinkage

Downloaded from acr.sagepub.com at BOSTON UNIV on October 18, 2015

Kaufmann et al.

1291

of the extracellular space secondary to resorption of plasma and changes in the conformation of the hemoglobin molecule. Other studies failed to demonstrate a benefit when using DWI as a discriminator between benign and malignant PVT (38). In any case, the application of DWI for response monitoring of HCC is limited to the subgroup of tumors presenting with restricted water diffusion in the baseline setting. During local or systemic therapy, signal intensity changes such as an increase on both T1W and T2W images as well as a decrease in contrast enhancement, reflect either hemorrhage or necrosis and need to be considered for correct evaluation of response to treatment (Fig. 8f–i) (40). The use of higher magnetic fields (7/9.4 Tesla) and the implementation of susceptibility-weighted imaging (SWI) are expected to further increase the sensitivity of MRI in this respect (41,42).

Conclusion Accurate diagnosis of benign or malignant portal vein infiltration is accomplished by most contrast-enhanced imaging techniques. However, cognizance of their strengths and limitations is mandatory. CEUS should represent the method of choice in the primary diagnosis of PVT in compliant patients. VPCT is less affected by motion artifacts and (just like dynamic contrastenhanced MRI) yields perfusion parameters that can be quantified and are therefore more suited for characterization and response monitoring studies. Conflict of interest None declared.

Funding This research received no specific grant from any funding agency in the public, commercial or not-for-profit sectors.

References 1. El-Serag HB. Epidemiology of hepatocellular carcinoma in USA. Hepatol Res 2007;37(Suppl. 2): S88–94. 2. El-Serag HB, Rudolph KL. Hepatocellular carcinoma: epidemiology and molecular carcinogenesis. Gastroenterology 2007;132:2557–2576. 3. Parkin DM, Bray F, Ferlay J, et al. Estimating the world cancer burden: Globocan 2000. Int J Cancer 2001;94: 153–156. 4. Sakata J, Shirai Y, Wakai T, et al. Preoperative predictors of vascular invasion in hepatocellular carcinoma. Eur J Surg Oncol 2008;34:900–905. 5. Takizawa D, Kakizaki S, Sohara N, et al. Hepatocellular carcinoma with portal vein tumor thrombosis: clinical characteristics, prognosis, and patient survival analysis. Dig Dis Sci 2007;52:3290–3295.

6. Minagawa M, Makuuchi M. Treatment of hepatocellular carcinoma accompanied by portal vein tumor thrombus. World J Gastroenterol 2006;12:7561–7567. 7. Zhou J, Tang ZY, Wu ZQ, et al. Factors influencing survival in hepatocellular carcinoma patients with macroscopic portal vein tumor thrombosis after surgery, with special reference to time dependency: a single-center experience of 381 cases. Hepatogastroenterology 2006; 53:275–280. 8. Kim KM, Kim JH, Park IS, et al. Reappraisal of repeated transarterial chemoembolization in the treatment of hepatocellular carcinoma with portal vein invasion. J Gastroenterol Hepatol 2009;24:806–814. 9. Zhao M, Wang JP, Li W, et al. [Comparison of safety and efficacy for transcatheter arterial chemoembolization alone and plus radiofrequency ablation in the treatment of single branch portal vein tumor thrombus of hepatocellular carcinoma and their prognosis factors]. Zhonghua Yi Xue Za Zhi 2011;91:1167–1172. 10. Cho JY, Paik YH, Park HC, et al. The feasibility of combined transcatheter arterial chemoembolization and radiotherapy for advanced hepatocellular carcinoma. Liver Int 2014;34:795–801. 11. Kurosaki I, Kawachi Y, Nihei K, et al. Liver perfusion chemotherapy with 5-Fluorouracil followed by systemic gemcitabine administration for resected pancreatic cancer: preliminary results of a prospective phase 2 study. Pancreas 2009;38:161–167. 12. Lau WY, Sangro B, Chen PJ, et al. Treatment for hepatocellular carcinoma with portal vein tumor thrombosis: the emerging role for radioembolization using yttrium-90. Oncology 2013;84:311–318. 13. Mizandari M, Ao G, Zhang Y, et al. Novel percutaneous radiofrequency ablation of portal vein tumor thrombus: safety and feasibility. Cardiovasc Intervent Radiol 2013; 36:245–248. 14. Tang QH, Li AJ, Yang GM, et al. Surgical resection versus conformal radiotherapy combined with TACE for resectable hepatocellular carcinoma with portal vein tumor thrombus: a comparative study. World J Surg 2013;37:1362–1370. 15. Yang M, Fang Z, Yan Z, et al. Transarterial chemoembolisation (TACE) combined with endovascular implantation of an iodine-125 seed strand for the treatment of hepatocellular carcinoma with portal vein tumour thrombosis versus TACE alone: a two-arm, randomised clinical trial. J Cancer Res Clin Oncol 2014;140:211–219. 16. Zhao Y, Cai G, Zhou L, et al. Transarterial chemoembolization in hepatocellular carcinoma with vascular invasion or extrahepatic metastasis: A systematic review. Asia Pac J Clin Oncol 2013;9:357–364. 17. Chung JW, Park JH, Han JK, et al. Hepatocellular carcinoma and portal vein invasion: results of treatment with transcatheter oily chemoembolization. Am J Roentgenol 1995;165:315–321. 18. Catalano OA, Choy G, Zhu A, et al. Differentiation of malignant thrombus from bland thrombus of the portal vein in patients with hepatocellular carcinoma: application of diffusion-weighted MR imaging. Radiology 2010; 254:154–162.

Downloaded from acr.sagepub.com at BOSTON UNIV on October 18, 2015

1292

Acta Radiologica 56(11)

19. Therasse P, Arbuck SG, Eisenhauer EA, et al. New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst 2000;92:205–216. 20. Ueno N, Kawamura H, Takahashi H, et al. Characterization of portal vein thrombus with the use of contrast-enhanced sonography. J Ultrasound Med 2006;25:1147–1152. 21. Hatanaka K, Kudo M, Minami Y, et al. Sonazoidenhanced ultrasonography for diagnosis of hepatic malignancies: comparison with contrast-enhanced CT. Oncology 2008;75(Suppl. 1): 42–47. 22. Raza SA, Jang HJ, Kim TK. Differentiating malignant from benign thrombosis in hepatocellular carcinoma: contrast-enhanced ultrasound. Abdom Imaging 2014;39: 153–161. 23. Tublin ME, Dodd GD, 3rd, Baron RL. Benign and malignant portal vein thrombosis: differentiation by CT characteristics. Am J Roentgenol 1997;168:719–723. 24. Shah ZK, McKernan MG, Hahn PF, et al. Enhancing and expansile portal vein thrombosis: value in the diagnosis of hepatocellular carcinoma in patients with multiple hepatic lesions. Am J Roentgenol 2007;188: 1320–1323. 25. Bota S, Piscaglia F, Marinelli S, et al. Comparison of international guidelines for noninvasive diagnosis of hepatocellular carcinoma. Liver Cancer 2012;1:190–200. 26. Bellomi M, Viotti S, Preda L, et al. Perfusion CT in solid body-tumours. Part II: Clinical applications and future development. Radiol Med 2010;115:858–874. 27. Petralia G, Fazio N, Bonello L, et al. Perfusion computed tomography in patients with hepatocellular carcinoma treated with thalidomide: initial experience. J Comput Assist Tomogr 2011;35:195–201. 28. Frampas E, Lassau N, Zappa M, et al. Advanced hepatocellular carcinoma: early evaluation of response to targeted therapy and prognostic value of perfusion CT and dynamic contrast enhanced ultrasound. Preliminary results. Eur J Radiol 2013;82:e205–211. 29. Sahani DV, Holalkere NS, Mueller PR, et al. Advanced hepatocellular carcinoma: CT perfusion of liver and tumor tissue–initial experience. Radiology 2007;243: 736–743. 30. Fattovich G, Stroffolini T, Zagni I, et al. Hepatocellular carcinoma in cirrhosis: incidence and risk factors. Gastroenterology 2004;127:S35–50.

31. Ippolito D, Capraro C, Casiraghi A, et al. Quantitative assessment of tumour associated neovascularisation in patients with liver cirrhosis and hepatocellular carcinoma: role of dynamic-CT perfusion imaging. Eur Radiol 2012;22:803–811. 32. Park JW, Kim JH, Kim SK, et al. A prospective evaluation of 18F-FDG and 11C-acetate PET/CT for detection of primary and metastatic hepatocellular carcinoma. J Nucl Med 2008;49:1912–1921. 33. Sun L, Guan YS, Pan WM, et al. Highly metabolic thrombus of the portal vein: 18F fluorodeoxyglucose positron emission tomography/computer tomography demonstration and clinical significance in hepatocellular carcinoma. World J Gastroenterol 2008;14:1212–1217. 34. Sun L, Guan YS, Pan WM, et al. Positron emission tomography/computer tomography in guidance of extrahepatic hepatocellular carcinoma metastasis management. World J Gastroenterol 2007;13:5413–5415. 35. Li C, Hu A, Haacke M, et al. Direct portal vein thrombosis visualization with t2*–weighted magnetic resonance imaging. Int J Med Sci 2013;10:1570–1574. 36. Kudo M. Hepatocellular carcinoma 2009 and beyond: from the surveillance to molecular targeted therapy. Oncology 2008;75(Suppl. 1): 1–12. 37. Granito A, Galassi M, Piscaglia F, et al. Impact of gadoxetic acid (Gd-EOB-DTPA)-enhanced magnetic resonance on the non-invasive diagnosis of small hepatocellular carcinoma: a prospective study. Aliment Pharmacol Ther 2013;37:355–363. 38. Sandrasegaran K, Tahir B, Nutakki K, et al. Usefulness of conventional MRI sequences and diffusion-weighted imaging in differentiating malignant from benign portal vein thrombus in cirrhotic patients. Am J Roentgenol 2013;201:1211–1219. 39. Maksimovic O, Schraml C, Hartmann JT, et al. Evaluation of response in malignant tumors treated with the multitargeted tyrosine kinase inhibitor sorafenib: a multitechnique imaging assessment. Am J Roentgenol 2010;194:5–14. 40. Horger M, Lauer UM, Schraml C, et al. Early MRI response monitoring of patients with advanced hepatocellular carcinoma under treatment with the multikinase inhibitor sorafenib. BMC Cancer 2009;9:208. 41. Fischer A, Kraff O, Maderwald S, et al. Non-enhanced t1-weighted liver vessel imaging at 7 tesla. PLoS One 2014;9:e97465. 42. Liu S, Mok K, Neelavalli J, et al. Improved MR venography using quantitative susceptibility-weighted imaging. J Magn Reson Imaging 2014;40:698–708.

Downloaded from acr.sagepub.com at BOSTON UNIV on October 18, 2015

Modern multimodality diagnosis of portal vein infiltration in hepatocellular carcinoma and expected changes during current therapies.

Diagnosis of malignant portal vein infiltration and especially differentiation from bland thrombosis is challenging for most imaging techniques. The u...
2MB Sizes 0 Downloads 4 Views