Hepatocellular Carcinoma: Targeted Therapy and Multidisciplinary P26 pdf

10 302 0
Hepatocellular Carcinoma: Targeted Therapy and Multidisciplinary P26 pdf

Đang tải... (xem toàn văn)

Thông tin tài liệu

14 Liver Transplant for Hepatocellular Carcinoma 235 51. Aloia TA, Fahy BN (2008) A decision analysis model predicts the optimal treatment path- way for patients with colorectal cancer and resectable synchronous liver metastases. Clin Colorectal Cancer 7:197–201 52. Lu DS, Yu NC, Raman SS et al (2005) Percutaneous radiofrequency ablation of hepatocellular carcinoma as a bridge to liver transplantation. Hepatology 41:1130–1137 53. Lu DS, Yu NC, Raman SS et al (2005) Radiofrequency ablation of hepatocellular carcinoma: treatment success as defined by histologic examination of the explanted liver. Radiology 234:954–960 54. Mazzaferro V, Battiston C, Perrone S et al (2004) Radiofrequency ablation of small hepatocel- lular carcinoma in cirrhotic patients awaiting liver transplantation: a prospective study. Ann Surg 240:900–909 55. Salem R, Thurston KG (2006) Radioembolization with yttrium-90 microspheres: a state- of-the-art brachytherapy treatment for primary and secondary liver malignancies: part 3: comprehensive literature review and future direction. J Vasc Interv Radiol 17:1571–1593 56. Kulik LM, Atassi B, van Holsbeeck L et al (2006) Yttrium-90 microspheres (TheraSphere) treatment of unresectable hepatocellular carcinoma: downstaging to resection, RFA and bridge to transplantation. J Surg Oncol 94:572–586 57. Riaz A, Kulik L, Lewandowski RJ et al (2009) Radiologic-pathologic correlation of hepato- cellular carcinoma treated with internal radiation using yttrium-90 microspheres. Hepatology 49:1185–1193 58. Lo CM, Liu CL, Chan SC et al (2007) A randomized, controlled trial of postoperative adjuvant interferon therapy after resection of hepatocellular carcinoma. Ann Surg 245:831–842 59. Simonetti RG, Liberati A, Angiolini C, Pagliaro L (1997) Treatment of hepatocellular carcinoma: a systematic review of randomized controlled trials. Ann Oncol 8:117–136 60. Abou-Alfa GK, Schwartz L, Ricci S et al (2006) Phase II study of sorafenib in patients with advanced hepatocellular carcinoma. J Clin Oncol 24:4293–4300 61. Llovet JM, Ricci S, Mazzaferro V et al (2008) Sorafenib in advanced hepatocellular carcinoma. N Engl J Med 359:378–390 62. Llovet JM, Bruix J (2008) Molecular targeted therapies in hepatocellular carcinoma. Hepatology 48:1312–1327 63. Fernandez JA, Robles R, Marin C et al (2003) Can we expand the indications for liver trans- plantation among hepatocellular carcinoma patients with increased tumor size? Transplant Proc 35:1818–1820 64. Leung JY, Zhu AX, Gordon FD et al (2004) Liver transplantation outcomes for early-stage hepatocellular carcinoma: results of a multicenter study. Liver Transpl 10:1343–1354 65. Yao FY, Ferrell L, Bass NM, Bacchetti P, Ascher NL, Roberts JP (2002) Liver transplanta- tion for hepatocellular carcinoma: comparison of the proposed UCSF criteria with the Milan criteria and the Pittsburgh modified TNM criteria. Liver Transpl 8:765–774 66. Mazzaferro V, Llovet JM, Miceli R et al (2009) Predicting survival after liver transplan- tation in patients with hepatocellular carcinoma beyond the Milan criteria: a retrospective, exploratory analysis. Lancet Oncol 10:35–43 67. Majno PE, Adam R, Bismuth H et al (1997) Influence of preoperative transarterial lipiodol chemoembolization on resection and transplantation for hepatocellular carcinoma in patients with cirrhosis. Ann Surg 226:688–701 68. Yao FY, Hirose R, LaBerge JM et al (2005) A prospective study on downstaging of hepatocellular carcinoma prior to liver transplantation. Liver Transpl 11:1505–1514 69. Llovet JM, Schwartz M, Fuster J, Bruix J (2006) Expanded criteria for hepatocellular car- cinoma through down-staging prior to liver transplantation: not yet there. Semin Liver Dis 26:248–253 70. Belghiti J, Cortes A, Abdalla EK et al (2003) Resection prior to liver transplantation for hepatocellular carcinoma. Ann Surg 238:885–893 71. Fong Y, Sun RL, Jarnagin W, Blumgart LH (1999) An analysis of 412 cases of hepatocellular carcinoma at a Western center. Ann Surg 229:790–800 236 T.A. Aloia et al. 72. Ercolani G, Grazi GL, Ravaioli M et al (2003) Liver resection for hepatocellular carcinoma on cirrhosis: univariate and multivariate analysis of risk factors for intrahepatic recurrence. Ann Surg 237:536–543 73. Adam R, Azoulay D, Castaing D et al (2003) Liver resection as a bridge to transplantation for hepatocellular carcinoma on cirrhosis: a reasonable strategy? Ann Surg 238:508–519 74. Kulik L, Abecassis M (2004) Living donor liver transplantation for hepatocellular carcinoma. Gastroenterology 127(5 Suppl 1):S277–S282 75. Fisher RA, Kulik LM, Freise CE et al (2007) Hepatocellular carcinoma recurrence and death following living and deceased donor liver transplantation. Am J Transplant 7: 1601–1608 76. Thuluvath PJ, Yoo HY (2004) Graft and patient survival after adult live donor liver transplan- tation compared to a matched cohort who received a deceased donor transplantation. Liver Transpl 10:1263–1238 77. Hwang S, Lee SG, Joh JW, Suh KS, Kim DG (2005) Liver transplantation for adult patients with hepatocellular carcinoma in Korea: comparison between cadaveric donor and living donor liver transplantations. Liver Transpl 11:1265–1272 78. Lo CM, Fan ST, Liu CL, Chan SC, Wong J (2004) The role and limitation of living donor liver transplantation for hepatocellular carcinoma. Liver Transpl 10:440–447 79. Sarasin FP, Majno PE, Llovet JM, Bruix J, Mentha G, Hadengue A (2001) Living donor liver transplantation for early hepatocellular carcinoma: A life-expectancy and cost-effectiveness perspective. Hepatology 33:1073–1079 80. Zavaglia C, De Carlis L, Alberti AB et al (2005) Predictors of long-term survival after liver transplantation for hepatocellular carcinoma. Am J Gastroenterol 100:2708–2716 81. Margarit C, Charco R, Hidalgo E, Allende H, Castells L, Bilbao I (2002) Liver t ransplantation for malignant diseases: selection and pattern of recurrence. World J Surg 26:257–263 82. Schlitt HJ, Neipp M, Weimann A et al (1999) Recurrence patterns of hepatocellular and fibrolamellar carcinoma after liver transplantation. J Clin Oncol 17:324–331 83. Shimoda M, Ghobrial RM, Carmody IC et al (2004) Predictors of survival after liver transplantation for hepatocellular carcinoma associated with Hepatitis C. Liver Transpl 10:1478–1486 84. Thuluvath PJ, Maheshwari A, Thuluvath NP, Nguyen GC, Segev DL (2009) Survival after liver transplantation for hepatocellular carcinoma in the model for end-stage liver disease and pre-model for end-stage liver disease eras and the independent impact of hepatitis C virus. Liver Transpl 15:754–762 85. Klintmalm GB (1998) Liver transplantation for hepatocellular carcinoma: a registry report of the impact of tumor characteristics on outcome. Ann Surg 228:479–490 86. Pawlik TM, Delman KA, Vauthey JN et al (2005) Tumor size predicts vascular invasion and histologic grade: Implications for selection of surgical treatment for hepatocellular carcinoma. Liver Transpl 11:1086–1092 87. Esnaola NF, Lauwers GY, Mirza NQ et al (2002) Predictors of microvascular invasion in patients with hepatocellular carcinoma who are candidates for orthotopic liver transplantation. J Gastrointest Surg 6:224–232 88. Busuttil RW, Farmer DG, Yersiz H et al (2005) Analysis of long-term outcomes of 3200 liver transplantations over two decades: a single-center experience. Ann Surg 241:905–918 89. Vivarelli M, Bellusci R, Cucchetti A et al (2002) Low recurrence rate of hepatocellular carcinoma after liver transplantation: better patient selection or lower immunosuppression? Transplantation 74:1746–1751 90. Vivarelli M, Cucchetti A, Piscaglia F et al (2005) Analysis of risk factors for tumor recurrence after liver transplantation for hepatocellular carcinoma: key role of immunosuppression. Liver Transpl 11:497–503 91. Guba M, von Breitenbuch P, Steinbauer M et al (2002) Rapamycin inhibits primary and metastatic tumor growth by antiangiogenesis: involvement of vascular endothelial growth factor. Nat Med 8:128–135 14 Liver Transplant for Hepatocellular Carcinoma 237 92. Aloia TA, Goss JA (2009) A report of outcomes after orthotopic liver transplant with allografts from heparin antibody-positive donors. Exp Clin Transplant 7:13–17 93. Guba M, Steinbauer M, Ruhland V et al (2002) Elevated MIA serum levels are predictors of poor prognosis after surgical resection of metastatic malignant melanoma. Oncol Reports 9:981–984 94. Schumacher G, Oidtmann M, Rosewicz S et al (2002) Sirolimus inhibits growth of human hepatoma cells in contrast to tacrolimus which promotes cell growth. Transplant Proc 34:1392–1393 95. Kneteman NM, Oberholzer J, Al Saghier M et al (2004) Sirolimus-based immunosuppression for liver transplantation in the presence of extended criteria for hepatocellular carcinoma. Liver Transpl 10:1301–1311 96. Marsh JW, Finkelstein SD, Demetris AJ et al (2003) Genotyping of hepatocellular carcinoma in liver transplant recipients adds predictive power for determining recurrence-free survival. Liver Transpl 9:664–671 97. UNOS/OPTN policy 3.6.4.4. http://www.optn.org. Accessed November 2009 Chapter 15 Vascular Resection for Hepatocellular Carcinoma Robin D. Kim and Alan W. Hemming Keywords Vascular resection · Vascular reconstruction · HCC · Intraoperative strategies for hepatic/vascular resections · Ante situm procedure Introduction Liver surgery has progressed over the last two decades to become a distinct area of specialization. Strategies such as portal vein embolization (Chapter 11)to induce growth of the planned liver remnant permit more aggressive resections, and improved imaging allows the surgeon to assess tumor position in relation to the intrahepatic vasculature. Liver transplantation has also progressed, but has been limited by the shortage of cadaveric donors. The development of live donor liver transplantation in response to this organ shortage has, in turn, led to techniques that can also be applied in non-transplant liver surgery. Resection and reconstruction of portal vein, hepatic artery, bile duct, and hepatic veins, all standard components of live donor liver transplantation, can be used in resecting complex HCC lesions by surgeons experienced in techniques developed for both liver resection and trans- plantation. Vascular resection and reconstruction is utilized to both achieve adequate oncologic tumor clearance and also preserve uninvolved hepatic parenchyma when vascular inflow or outflow is involved. In this chapter, we examine the role and techniques of vascular resection and reconstruction for HCC. A.W. Hemming (B) Division of Transplantation and Hepatobiliary Surgery, Department of Surgery, University of California, San Diego, CA, USA 239 K.M. McMasters, J N. Vauthey (eds.), Hepatocellular Carcinoma, DOI 10.1007/978-1-60327-522-4_15, C  Springer Science+Business Media, LLC 2011 240 R.D. Kim and A.W. Hemming The Pathophysiology of Vascular Invasion in HCC Vascular invasion is an important characteristic of HCC as not only may it require vascular resection/reconstruction, but it is also a significant predictor of recur- rence following resection [1, 2]. In one series of 322 patients who underwent curative resection for HCC, macroscopic vascular invasion (that which is visible on gross section) and microscopic vascular invasion (that which is detected at histology) were detected in 15.5% and 59.0%, respectively [2]. The pathophysiol- ogy of vascular invasion has been elucidated by histologic studies of early cases. Immunohistochemistry studies of tumors using CD31, a marker for endothelial cells, show that in most cases invasion begins when tumor nests surrounded by sinusoidal vessels extend into the portal and hepatic veins. These endothelial-coated tumor emboli enter the circulation, adhere to local tributaries and proliferate, and can embolize to distant sites once entering the systemic circulation [3]. This mecha- nism is invasion independent as it does not depend on the invasive activity of tumor cells but rather a relationship with endothelial progenitors [4]. This intravasation of tumor nests by tumor vessels explains the shunting seen in the arterial phase of CT scans, as the contrast that is delivered arterially is shunted to neighboring hepatic or portal veins through a well-vascularized tumor (Fig. 15.1). Fig. 15.1 Arterial phase CT demonstrating arterial flow within the portal vein, indicating vascularized tumor within the portal vein Hepatocellular cancer invades either portal or hepatic veins by local extension through the above mechanism, ultimately occluding and expanding the vascular space. In portal vein invasion, tumor cells may disseminate into distal branches resulting in intrahepatic metastases. This event is responsible for the tumor satellito- sis that accompanies a dominant tumor within the same segment. Others have used radiopaque injection of tumors to confirm that the portal vein may act as an efferent vessel in the setting of portal hypertension, thus explaining the tumor emboli found 15 Vascular Resection for Hepatocellular Carcinoma 241 in the rectal veins and esophageal varices in autopsy studies [3, 5]. One large series of 1023 patients who underwent resections for HCC found macroscopic portal vein tumor thrombus in 54 patients (5.4%) [6]. In hepatic vein invasion, the tumor may grow and extend into the inferior vena cava and the right atrium, and when large may create an arterio-venous shunt bypassing the sinusoidal filter. Tumor dissemination from hepatic vein invasion is significant when cells are released as multicellular tumor nests with preserved cell–cell and cell–matrix interactions [7]. Unlike single cells, these clusters can survive anoikis, mechanical disruption, and host defenses to metastasize distant sites such as the lungs [8]. Some of the risk factors of vascular invasion in HCC include tumor size [2, 9, 10], tumor number, [9, 11], histologic grade [9, 10, 12], and elevated alpha-fetoprotein level [2, 11]. It is essential to understand the pathophysiology that accompanies the need for vascular resection in HCC and weigh the risk of recurrence against the increased technical demands and risks of any proposed procedure. Evaluation and Work-Up of the Patient with HCC for Resection Underlying Liver Disease in the Patient with HCC A careful assessment of the patients underlying liver function is needed to determine the operative risk liver failure and death following resection for HCC (Chapter 9). In general, Child–Pugh class C is a contraindication to resection, and early class B patients without portal hypertension may undergo minor resections from wedge resection to a single segmentectomy. Child–Pugh class A patients that are consid- ered for major hepatectomy (resection of four or more segments) should undergo assessment of both liver and physiologic status [13, 14]. In addition, strategies such as pre-operative portal vein embolization (PVE) to increase the future liver remnant (FLR) have been associated with decreased complications and extended surgical options for HCC patients (Chapter 11)[15–18]. Other risk factors are associated with liver failure and death following resec- tion of HCC. Portal hypertension (PH) is a contraindication to liver resection as it has been associated with increased morbidity and mortality following major resection [19]. PH is defined as a hepatic vein pressure gradient (HVPG) greater than 10 mm Hg, and some associated signs include esophageal varices, anatomic portosystemic shunts, and ascites [20]. Thrombocytopenia with platelet counts <100,000 cells/mm 2 has been associated with portal hypertension and an increased in-hospital mortality following liver resection [13]. There are occasional cases in which portal hypertension exists in the setting of a non-cirrhotic liver secondary to either partial portal vein or hepatic vein occlusion by tumor. In rare instances resection can be considered in these situations if the surgeon is convinced that the underlying liver is non-cirrhotic with the assumption that portal pressures and liver function will improve after resection and the mechanical problem caused by tumor obstruction is relieved. Active viral hepatitis is another risk factor for 242 R.D. Kim and A.W. Hemming liver failure and death following HCC resection and is suggested by serum alanine aminotransferase levels (fourfold in one series) [21, 22]. For major hepatectomies that involve vascular resection, particularly of the hepatic veins and inferior vena cava (IVC), it will be the rare patient that has any sig- nificant degree of cirrhosis that would be considered for resection. Cirrhotic patients with portal vein involvement requiring tumor thrombectomy or portal vein resection that otherwise meet standard resection criteria can be considered for resection. Pre-operative Imaging Pre-operative imaging is required to stage the tumor, to assess its position in relation to hepatic vasculature, and to plan the liver resection to achieve an R0 resection while preserving adequate liver remnant. Accurate imaging of the intrahepatic architecture enhanced with three-dimensional reconstruction is important to assess the possible need for vascular reconstruction. For example, imaging may clarify the venous anatomy in the setting where a tumor in segment 7 or 8 requires sacrifice of the main right hepatic vein and yet segment 6 requires preservation. Accurate imaging may indicate the need for or obviate vascular reconstruction such as a large inferior hepatic vein draining segment 6 that makes reconstruction of the main right hepatic vein unnecessary. Alternatively, anatomy may be discovered that requires vascular reconstruction such as a large segment 6 tributary to the middle hepatic vein that would require reconstruction in an extended left hepatectomy [23]. Triphasic spiral computed tomography (CT) of the liver with concurrent assess- ment of the chest is t he most widely used modality for planning surgery and staging for HCC. The classic appearance of a lesion hyper-enhancing on the arterial phase with subsequent hypo-enhancement (“washout”) in the portal venous phase in the setting of underlying liver disease is diagnostic, with some variability depending on size, location, and level of fat or fibrosis of the surrounding liver. In addition, gross vascular invasion can be visualized by local extension and expansion of tumor thrombus in a hepatic or portal vein from the tumor and by the shunting of contrast through the thrombus during the arterial phase (Fig. 15.1). Despite its popularity, CT is limited in detecting macroscopic vascular invasion as the findings may be subtle. One group has reported that CT scan detected 68% of portal vein thrombi associ- ated with HCC and correctly characterized 68% as malignant following pathologic examination [24]. Magnetic resonance imaging (MRI) with gadolinium for MR angiography and venography may further demonstrate the hepatic veins, particularly when all three are involved resulting in outflow obstruction that prevents adequate flow of contrast into the hepatic veins during CT. In addition, MRI with agents such as super- magnetic iron oxide has been shown to detect even microvascular invasion with sensitivity, specificity, and accuracy rates of 82, 84, and 86%, respectively [25]. Three-dimensional reconstruction of axial images and volumetric assessment of the total and planned liver remnant have been found to be more accurate than 15 Vascular Resection for Hepatocellular Carcinoma 243 axial imaging before liver resection [26, 27]. These imaging techniques coupled with computer-aided functional remnant predictions, based on not only spared liver parenchyma but also simulated inflow/outflow changes, have been shown to not only alter surgery extent but also the need for vascular reconstruction [28]. Although a remnant liver volume of 25% after resection is generally adequate in the uninjured liver, extended liver resections for HCC generally occur in the setting of compro- mised liver due to fibrosis. If the FLR is projected to be less that 40%, pre-operative PVE can be used to increase the size of the liver remnant. In addition, with vascular reconstruction and the possible use of cold perfusion the liver receives an additional ischemic injury beyond that of standard liver resection. We have arbitrarily chosen to use PVE in any patient requiring major hepatectomy with vascular reconstruction who has an FLR <40% even in the setting of normal hepatic parenchyma. The lack of adequate growth following PVE is a sign of severe liver injury and inadequate regenerative capacity which precludes extended resection [17]. Intraoperative Strategies for Hepatic/Vascular Resections Both low central venous pressure (CVP) and inflow occlusion (Pringle maneuver) are strategies used in standard liver resections to minimize blood loss [29]. Low central venous pressure (CVP <6 mm Hg) can be achieved by positioning the patient in reverse Trendelenburg position [30], fluid restriction, diuresis and vasodilators. Low CVP during parenchymal transection decreases back-bleeding from the hepatic veins and their tributaries and is useful when extensive dissection of the hepatic veins is needed in preparation for vascular resection. Inflow occlusion (Pringle maneuver) decreases blood loss during liver transec- tion and is achieved by occluding the hepatic artery and portal vein using a large atraumatic vascular clamp or tourniquet. Although normal livers can tolerate up to 60 min of continuous inflow occlusion/warm ischemia [31], injured livers (from cirrhosis, biliary obstruction, or chemotherapy) tolerate significantly less ischemia before irreversible injury ensues [32]. Intermittent inflow occlusion for 15 min with 5 min breaks has been suggested to reduce liver injury [33]andmaybeusedin complex resections in which the parenchymal transection may be prolonged or when hepatic venous reconstruction are required at the completion of the liver transection. Despite these efforts, these complex cases are particularly at risk for ischemic injury as they are associated with underlying liver injury, greater blood loss at surgery, and longer periods of ischemia during vascular resection/reconstructions. Ischemic preconditioning (IP) has been suggested by some to protect the liver from subsequent ischemic injury [34]. Ischemic preconditioning is performed by applying the Pringle maneuver for 10 min and then reperfusing the liver for at least 10 min prior to reapplying inflow occlusion for the liver transection. The mechanisms by which ischemic preconditioning protects the liver include the upreg- ulation of the protective signals such as IL-6 and STAT3, alterations in energy metabolism, and abrogation of injurious events such as neutrophil accumulation, 244 R.D. Kim and A.W. Hemming microcirculatory disturbance, and reactive oxygen species, and proinflammatory mediators [35]. The benefits of IP for liver resections have not been found consis- tently. A recent meta-analysis has shown that in non-cirrhotic patients, IP before liver resection was associated with a decrease in blood transfusions but did not change mortality, liver failure, morbidity, or length of stay [36]. Although we use both the Pringle maneuver (15 min on, 5 min off) and ischemic preconditioning when necessary, our standard practice is to use no inflow occlusion at all during the hepatic parenchymal transection phase of the procedure if vascular reconstruc- tion is planned. Ideally the hepatic transection is done preserving perfusion to the remnant liver until the time that blood flow must be interrupted to resect and reconstruct the involved vessel. This minimizes the ischemic time and reduces liver injury. Technical Considerations for Hepatic Resection with Vascular Reconstruction and Published Experience Lesions Involving the Hilar Vessels Tumors of all types may involve the hilar vessels by extrinsic compression, and in the case of adenocarcinomas may invade from the outside. Hepatocellular cancer behaves in a different fashion. Although HCC may cause extrinsic compression due to size, this process rarely leads to invasion of the vessel wall. Instead, HCC has a propensity to invade nearby portal veins by extending tumor thrombi into sinusoids, then into the portal vein branch lumen itself, and this thrombus may extend to the ipsilateral, then main and contralateral portal veins. Generally, the hilar vessels are addressed after liver mobilization and ultra- sonography and before parenchymal transection to minimize blood loss. When pre-operative and intraoperative imaging rule out tumor thrombus in the case of extrinsic compression, the portal vein can be separated from the compressed hepatic parenchyma surrounding the tumor and still obtain adequate though small margins. However, when a tumor thrombus involves the main trunk of the right or left portal vein or extends down into the main portal vein or across to the contralateral por- tal vein, then proximal and distal control must be achieved without amputating the thrombus and creating a tumor embolus. The main and contralateral portal veins are dissected out well beyond tumor to prevent amputation and embolization of tumor at the time of clamp placement. The ipsilateral portal vein can be transected and the tumor thrombus extracted in most cases. The proximal and distal remnant portal veins are then flushed, and the ipsilateral stump closed. In cases where the tumor is adherent to the vein wall, the section of portal vein can be resected and in general a primary end-to-end anastomosis performed between the main portal vein to right or left branch (Fig. 15.2a–c). Up to 2 cm of vein can be resected without the use of a graft. Blood flow to the liver is maintained through the hepatic artery, and the portal vein clamp time is short. Although portal vein resections and reconstructions . Molecular targeted therapies in hepatocellular carcinoma. Hepatology 48:1312–1327 63. Fernandez JA, Robles R, Marin C et al (2003) Can we expand the indications for liver trans- plantation among hepatocellular. (TheraSphere) treatment of unresectable hepatocellular carcinoma: downstaging to resection, RFA and bridge to transplantation. J Surg Oncol 94:572–586 57. Riaz A, Kulik L, Lewandowski RJ et al (2009) Radiologic-pathologic. al (2007) Hepatocellular carcinoma recurrence and death following living and deceased donor liver transplantation. Am J Transplant 7: 1601–1608 76. Thuluvath PJ, Yoo HY (2004) Graft and patient

Ngày đăng: 07/07/2014, 09:20

Tài liệu cùng người dùng

  • Đang cập nhật ...

Tài liệu liên quan