• No results found

Towards safer liver resections - Chapter 1: General introduction and outline of thesis

N/A
N/A
Protected

Academic year: 2021

Share "Towards safer liver resections - Chapter 1: General introduction and outline of thesis"

Copied!
9
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

UvA-DARE is a service provided by the library of the University of Amsterdam (https://dare.uva.nl)

UvA-DARE (Digital Academic Repository)

Towards safer liver resections

Hoekstra, L.T.

Publication date

2012

Link to publication

Citation for published version (APA):

Hoekstra, L. T. (2012). Towards safer liver resections.

General rights

It is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), other than for strictly personal, individual use, unless the work is under an open content license (like Creative Commons).

Disclaimer/Complaints regulations

If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material inaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letter to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You will be contacted as soon as possible.

(2)

Chapter

General introduction

(3)
(4)

General introduction

Liver resection is the most effective treatment for primary or metastatic liver tumors.1,2

In patients with malignant hepatic tumors, fewer than 15-20% are suitable for surgical resection. Whereas extended liver resections are increasingly indicated because of advanced tumor in these patients, the extent of liver resection is restricted by the minimum volume of the liver remnant required to provide sufficient postoperative liver function. When patients are considered unresectable because of too small future remnant liver (FRL), neoadjuvant therapy or the combination of resection with local ablative techniques such as RFA may be applied. Alternatively, preoperative portal vein embolization (PVE) is an option to increase FRL volume through induction of regeneration of the hepatocellular mass of the non-embolized FRL.3,4 Following occlusion of the right or left branch of

the portal vein, atrophy of the embolized liver segments occurs, while hypertrophy of the contralateral, non-embolized liver lobe is induced.5,6 Liver regeneration is set off

in the contralateral liver lobe via a complex interaction of cytokines, growth factors and metabolic networks.7 The degree of hypertrophy after PVE is variable, especially in

patients with compromised livers.4 PVE is a relatively new intervention and is mostly used

in specialized centres. Numbers reported in clinical studies are relatively limited for this reason. PVE has shown to reduce the risk of liver failure3 and consequently increases the

number of patients who are able to undergo liver resection.

There is increasing evidence that PVE not only stimulates growth of the FRL but also accelerates tumor proliferation.7 Tumor progression after PVE creates a dilemma in terms

of optimal waiting time until resection.4 Surgery is usually performed 3-6 weeks after PVE

when sufficient increase of the FRL is considered to have occurred to allow a safe liver resection.8-10 Several studies reported tumor growth after PVE in the embolized and

non-embolized liver segments11-14 within the waiting time for resection under the influence

of the above mentioned cytokines and growth factors. In addition, since the liver has a dual blood supply by the portal vein and the hepatic artery, PVE will also result in a compensatory increase in hepatic arterial flow15 which further enhances tumor growth.

The challenge for future use of PVE is to limit the growth of tumor while efficiently inducing hypertrophy of the non-embolized liver lobe. Several strategies can be devised to limit post-PVE tumor progression, as are discussed in this thesis.

The numbers of hepatic resections have increased as tumors initially considered unresectable have become potentially resectable after preoperative or intraoperative interventions. Strategies such as PVE or two-stage resection16 rely heavily on the

tremendous regenerative ability of the normal liver. However, many of the patients who are candidates for resection also have impaired liver function due to fibrosis/cirrhosis, steatosis or chemotherapy induced parenchymal injury. These patients in particular, are at risk when undergoing extensive liver resection and all efforts should be directed to reducing postoperative complications.

General introduction

11

(5)

This thesis deals with the available options to perform safer liver resections. In part 1, several clinical questions regarding PVE, are adressed in an experimental model of PVE in the rabbit. Clinical studies mainly focus on liver regeneration markers, and the influence of PVE on tumor growth. In part 2, the aim is to assess complications after liver resection and to discuss methods and surgical approaches to prevent the most common complications.

Outline of the thesis

Part I

Postoperative liver failure is the major cause of mortality and morbidity after liver resection, and develops as a result of insufficient remnant liver function.17 Assessment

of liver function is therefore crucial in the preoperative work-up of patients who require (extensive) liver resection. Chapter 2 describes the physiological basis of the most frequently employed clinical liver function tests.

The extent of liver resection is restricted by the volume of the future remnant liver. Patients are considered resectable when the FRL is larger than 25-30% in patients with normal liver parenchyma, whereas a limit of 40% is taken into account in patients with diseased livers.4,7,18,19 One way to increase the FRL preoperatively is the use of PVE, which

has been clinically introduced in 1990 by Makuuchi et al.20 In chapter 3, the experiences

and outcomes of PVE and extensive resection in predamaged livers in our center are reported.

Although PVE is largely applied worldwide, many questions concerning the hypertrophy response and liver regeneration still need to be elucidated.4 Since liver

regeneration after PVE is variable, we evaluated several possible predictors of liver growth. In chapter 4 and 5, plasma bile salt levels, triglycerides, and ApoA-V were investigated in the prediction of the hypertrophy response after PVE in a rabbit PVE-model, as well as in a series of patients undergoing PVE. Chapter 6 deals with the role of thrombocytes in volumetric regeneration after portal vein embolization in a series of patients undergoing subsequent liver resection.

There is increasing evidence that PVE not only stimulates growth of the FRL but also increases tumor size as a result of cytokines, growth factors and an increased arterial blood supply to the tumor4,7,11,13,14,21-24, but the exact mechanisms of this phenomenon

are largely unknown. Growth of tumor may be accelerated, while micrometastases in the non-embolized remnant liver may also develop or progress. The rate of tumor growth after PVE had been examined in a series of patients in chapter 7.

Another strategy in conjunction with PVE is to selectively embolize the hepatic artery (HAE) branches to the tumor-bearing liver segments prior to PVE. HAE alone does not result in the desired atrophy-hypertrophy response. The combination of HAE and PVE, however, will result in hypertrophy of the non-embolized lobe, at the same time limiting further tumor growth induced by compensatory hyperperfusion of the hepatic artery.

(6)

Simultaneous embolization of the portal vein and hepatic artery obviously carries a high risk of parenchymal necrosis as a result of complete occlusion of the dual blood supply.25,26

The risk of necrosis is diminished when the hepatic artery and portal vein are embolized in sequential order. The optimal time-interval between embolization of the portal vein and hepatic artery is, however, unknown. Therefore, the optimal timing of portal vein embolization and transarterial (chemo)embolization is examined in a review presented in chapter 8.

Besides clinical studies that reported tumor growth acceleration after PVE, there is a need for optimization of treatment strategies to prevent tumor progression after portal vein embolization. Experimental studies are necessary to unravel several important clinical questions. We have developed a rabbit model in which PVE can be assessed using the same imaging methods used to evaluate patients after PVE.27 The combination of a VX2

liver tumor in this rabbit model allowed us to investigate tumor growth after PVE in a rabbit VX2 tumor model (chapter 9).

Ascites is a common complication after liver resection.28 It may contribute to liver

failure when large intra-abdominal ascitic fluid collections develop. Because little has been published about this troublesome complication after liver resection, we investigated in chapter 10, the incidence of ascites after hepatectomy with or without preoperative portal vein embolization, in addition to predictive factors for the development of post-resectional ascites.

Part II

Excessive blood loss during transection of the liver parenchyma is associated with adverse postoperative outcomes, which may culminate into liver failure especially when a small liver remnant is involved.29 To combat blood loss during liver resection, various methods

of hepatic inflow or simultaneous in- and outflow occlusion techniques have been introduced. A systematic literature search was conducted to update the effects of liver in- and outflow occlusion techniques during liver resection in chapter 11, focusing on blood loss and hepatic ischemia-reperfusion injury.

Biliary leakage after liver resection continues to be reported.30,31 Management of

bile leakage has changed in recent years, with to date, non-surgical procedures as the preferred treatment. The outcomes of biliary leakage and management were assessed in 381 patients who had undergone liver resection between 2005 and 2011, and the results are presented in chapter 12.

Within the field of liver surgery, the use of laparoscopy has increased substantially in recent years.32 Laparoscopic operations require insufflation of the abdominal cavity

(pneumoperitoneum, PP) with carbon dioxide gas to achieve adequate surgical exposure for instrumentation and surgical manoeuvres. Pneumoperitoneum obviously produces elevated intra-abdominal pressure with continuous compression of intra-abdominal organs which potentially influences hepatic microcirculatory perfusion. The study in chapter 13 was undertaken to investigate the influence of prolonged PP on liver function and hepatic

General introduction

13

(7)

microcirculatory parameters in a clinically relevant, porcine model of extended abdominal insufflation.

There is a prominent increase in laparoscopic approaches for primary colorectal carcinoma in recent years. Laparoscopy however, is only used in a selected group of patients with colorectal liver metastases in the Netherlands. There is much discussion in performing colonic and liver resections simultaneously. The aim of the study reported in chapter 14 was to evaluate our initial experiences of combined laparoscopic resection of colorectal cancer and synchronic hepatic metastases.

Staging laparoscopy (SL) has been found useful in determining appropriate treatment in several malignant tumors33-39, but is not regularly performed in patients

with hepatocellular carcinoma (HCC). SL may change treatment strategy, preventing unnecessary open exploration. The aim of the study in chapter 15 was to assess the value and outcomes of diagnostic laparoscopy in the treatment strategy for HCC.

Finally, in chapter 16 the results of the studies performed in this thesis are summarized and discussed.

(8)

References

1. Dinant S, de Graaf W, Verwer BJ et al. Risk assessment of posthepatectomy liver failure using hepatobiliary scintigraphy and CT volumetry. J Nucl Med 2007; 48:685-692.

2. Lehnert T, Otto G, Herfarth C. Therapeutic modalities and prognostic factors for primary and secondary liver tumors. World J Surg 1995; 19:252-263.

3. Abulkhir A, Limongelli P, Healey AJ et al. Preoperative portal vein embolization for major liver resection: a meta-analysis. Ann Surg 2008; 247:49-57.

4. van Gulik TM, van den Esschert JW, de Graaf W et al. Controversies in the use of portal vein embolization. Dig Surg 2008; 25:436-444.

5. Takayasu K, Muramatsu Y, Shima Y et al. Hepatic lobar atrophy following obstruction of the ipsilateral portal vein from hilar cholangiocarcinoma. Radiology 1986; 160:389-393.

6. Taub R. Liver regeneration: from myth to mechanism. Nat Rev Mol Cell Biol 2004; 5:836-847. 7. de Graaf W, van den Esschert JW, van Lienden KP et al. Induction of tumor growth after

preoperative portal vein embolization: is it a real problem? Ann Surg Oncol 2009; 16:423-430. 8. Abdalla EK, Hicks ME, Vauthey JN. Portal vein embolization: rationale, technique and future

prospects. Br J Surg 2001; 88:165-175.

9. de BT, Roche A, Vavasseur D et al. Portal vein embolization: utility for inducing left hepatic lobe hypertrophy before surgery. Radiology 1993; 188:73-77.

10. de Graaf W, van Lienden KP, van den Esschert JW et al. Increase in future remnant liver function after preoperative portal vein embolization. Br J Surg 2011; 98:825-834.

11. Barbaro B, Di SC, Nuzzo G et al. Preoperative right portal vein embolization in patients with metastatic liver disease. Metastatic liver volumes after RPVE. Acta Radiol 2003; 44:98-102. 12. Elias D, de BT, Roche A et al. During liver regeneration following right portal embolization the

growth rate of liver metastases is more rapid than that of the liver parenchyma. Br J Surg 1999; 86:784-788.

13. Hayashi S, Baba Y, Ueno K et al. Acceleration of primary liver tumor growth rate in embolized hepatic lobe after portal vein embolization. Acta Radiol 2007; 48:721-727.

14. Kokudo N, Tada K, Seki M et al. Proliferative activity of intrahepatic colorectal metastases after preoperative hemihepatic portal vein embolization. Hepatology 2001; 34:267-272.

15. Nagino M, Nimura Y, Kamiya J et al. Immediate increase in arterial blood flow in embolized hepatic segments after portal vein embolization: CT demonstration. AJR Am J Roentgenol 1998; 171:1037-1039.

16. Bowers KA, O’Reilly D, Bond-Smith GE et al. Feasibility study of two-stage hepatectomy for bilobar liver metastases. Am J Surg 2011.

17. van den Broek MA, Olde Damink SW, Dejong CH et al. Liver failure after partial hepatic resection: definition, pathophysiology, risk factors and treatment. Liver Int 2008; 28:767-780.

18. de BT, Roche A, Elias D et al. Preoperative portal vein embolization for extension of hepatectomy indications. Hepatology 1996; 24:1386-1391.

19. Kubota K, Makuuchi M, Kusaka K et al. Measurement of liver volume and hepatic functional reserve as a guide to decision-making in resectional surgery for hepatic tumors. Hepatology 1997; 26:1176-1181.

20. Makuuchi M, Thai BL, Takayasu K et al. Preoperative portal embolization to increase safety of major hepatectomy for hilar bile duct carcinoma: a preliminary report. Surgery 1990; 107:521-527. 21. Azoulay D, Raccuia JS, Castaing D et al. Right portal vein embolization in preparation for major

hepatic resection. J Am Coll Surg 1995; 181:266-269.

22. Elias D, Ouellet JF, de BT et al. Preoperative selective portal vein embolization before hepatectomy for liver metastases: long-term results and impact on survival. Surgery 2002; 131:294-299. 23. Pamecha V, Levene A, Grillo F et al. Effect of portal vein embolisation on the growth rate of

colorectal liver metastases. Br J Cancer 2009; 100:617-622.

General introduction

15

(9)

24. Ribero D, Abdalla EK, Madoff DC et al. Portal vein embolization before major hepatectomy and its effects on regeneration, resectability and outcome. Br J Surg 2007; 94:1386-1394.

25. Nakao N, Miura K, Takahashi H et al. Hepatocellular carcinoma: combined hepatic, arterial, and portal venous embolization. Radiology 1986; 161:303-307.

26. Yamakado K, Hirano T, Kato N et al. Hepatocellular carcinoma: treatment with a combination of transcatheter arterial chemoembolization and transportal ethanol injection. Radiology 1994; 193:75-80.

27. de Graaf W, van den Esschert JW, van Lienden KP et al. A rabbit model for selective portal vein embolization. J Surg Res 2011; 171:486-494.

28. Ishizawa T, Hasegawa K, Kokudo N et al. Risk factors and management of ascites after liver resection to treat hepatocellular carcinoma. Arch Surg 2009; 144:46-51.

29. Jarnagin WR, Gonen M, Fong Y et al. Improvement in perioperative outcome after hepatic resection: analysis of 1,803 consecutive cases over the past decade. Ann Surg 2002; 236:397-406. 30. Capussotti L, Ferrero A, Vigano L et al. Bile leakage and liver resection: Where is the risk? Arch

Surg 2006; 141:690-694.

31. Tanaka S, Hirohashi K, Tanaka H et al. Incidence and management of bile leakage after hepatic resection for malignant hepatic tumors. J Am Coll Surg 2002; 195:484-489.

32. Buell JF, Cherqui D, Geller DA et al. The international position on laparoscopic liver surgery: The Louisville Statement, 2008. Ann Surg 2009; 250:825-830.

33. Connor S, Barron E, Wigmore SJ et al. The utility of laparoscopic assessment in the preoperative staging of suspected hilar cholangiocarcinoma. J Gastrointest Surg 2005; 9:476-480.

34. de Graaf GW, Ayantunde AA, Parsons SL et al. The role of staging laparoscopy in oesophagogastric cancers. Eur J Surg Oncol 2007; 33:988-992.

35. Ellsmere J, Mortele K, Sahani D et al. Does multidetector-row CT eliminate the role of diagnostic laparoscopy in assessing the resectability of pancreatic head adenocarcinoma? Surg Endosc 2005; 19:369-373.

36. Kapiev A, Rabin I, Lavy R et al. The role of diagnostic laparoscopy in the management of patients with gastric cancer. Isr Med Assoc J 2010; 12:726-728.

37. Muntean V, Mihailov A, Iancu C et al. Staging laparoscopy in gastric cancer. Accuracy and impact on therapy. J Gastrointestin Liver Dis 2009; 18:189-195.

38. Stefanidis D, Grove KD, Schwesinger WH et al. The current role of staging laparoscopy for adenocarcinoma of the pancreas: a review. Ann Oncol 2006; 17:189-199.

39. Weber SM, DeMatteo RP, Fong Y et al. Staging laparoscopy in patients with extrahepatic biliary carcinoma. Analysis of 100 patients. Ann Surg 2002; 235:392-399.

Referenties

GERELATEERDE DOCUMENTEN

However, the chemistry o f lanthanide and group 4 metal organometallic complexes bearing donor functionalized alkoxide ligands has not yet been well explored,

The work described herein offers new insights into stable radical chemistry by presenting a detailed study of the magnetic and redox properties of novel 6-oxoverdazyl radicals

If the intersystem crossing quantum yield was assumed to be the same for the annelated CPDs as was measured for the simple substituted DMDHPs (i.e. 10'^) then

More specifically, chapter two outlines the systematic study of protonation and hydrogen bonding effects on verdazyl electrochemistry, while chapter three investigates the

Reactions of 5.1 with Ru(hfac) 2 (MeCN) 2 generates two chemically similar and isolable compounds. The solid state structures of both compounds suggest the ligand

In this section, we apply selected MCL-containing copolymers to compare the structure and drug delivery function of PAX-loaded PNPs prepared using the conventional approach of

Electron withdrawing substituent (NO 2 ) at the para positions of the internal styryl groups accelerate, whereas electron donating groups (MeO, Me) decelerate the thermal

A simple three-link biomechanical model (referred to as the “plant”), commonly used in sit-to-stand analysis, was used to simulate the move- ment. The aim of this work is to