11.02.2015 Views

barcelona . spain - European Association for the Study of the Liver

barcelona . spain - European Association for the Study of the Liver

barcelona . spain - European Association for the Study of the Liver

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

BARCELONA . SPAIN<br />

152 POSTGRADUATE COURSE SYLLABUS ALCOHOLIC LIVER DISEASE 153<br />

APRIL 18 - 19/2012 THE INTERNATIONAL LIVER CONGRESS TM 2012<br />

STEM CELLS IN ALCOHOLIC LIVER DISEASE: THE ROAD FROM<br />

ANIMAL MODELS TO HUMAN STUDIES<br />

Laurent Spahr<br />

Geneva, Switzerland<br />

E-mail: Laurent.Spahr@hcuge.ch<br />

Grants: Supported by <strong>the</strong> Clinical Research Center, University Hospital and Faculty <strong>of</strong> Medicine, Geneva,<br />

<strong>the</strong> Louis-Jeantet Foundation, and Foundation <strong>for</strong> <strong>Liver</strong> and Gut Studies (FLAGS) in Geneva<br />

KEY POINTS<br />

• The impaired regenerative capacity <strong>of</strong> alcoholic cirrhosis with ASH (acute-on chronic ALD)<br />

participates to <strong>the</strong> high morbidity and mortality <strong>of</strong> <strong>the</strong> disease.<br />

• Under specific experimental conditions, animal studies show that bone marrow (BM)-derived<br />

stem cells participate in liver regeneration, but mechanisms are incompletely elucidated.<br />

• Some benefits derived from BM-derived cell <strong>the</strong>rapy seem associated with <strong>the</strong> co-administration<br />

<strong>of</strong> G-CSF to mobilize hematopoietic stem cells, produce humoral factors, and facilitate cells<br />

homing to <strong>the</strong> site <strong>of</strong> injury.<br />

• In patients with cirrhosis and ASH, G-CSF stimulates <strong>the</strong> short-term proliferation <strong>of</strong> hepatic<br />

progenitors.<br />

• In patients with decompensated liver disease <strong>of</strong> mixed aetiology, results <strong>of</strong> <strong>the</strong> few RCTs<br />

suggest that autologous BM stem cell transplantation is safe and may transiently improve liver<br />

function in non alcoholic cirrhosis.<br />

• Autologous BM stem cell transplantation in decompensated ALD doesn’t improve liver function<br />

(MELD score) over a 3-month period.<br />

• Future studies should better characterize <strong>the</strong> efficiency <strong>of</strong> G-CSF and BM stem cell <strong>the</strong>rapy in<br />

<strong>the</strong> regeneration and repair <strong>of</strong> decompensated alcoholic liver, and explore <strong>the</strong> fate and potential<br />

<strong>of</strong> transplanted cells.<br />

INTRODUCTION<br />

Cirrhosis is associated with insufficient regeneration in response to injury, due to multiples causes including<br />

extensive fibrosis, inflammation, steatosis and replicative senescence <strong>of</strong> hepatocytes, all <strong>of</strong> which constitute<br />

an unsuitable environment <strong>for</strong> liver cell proliferation. Acute-on-chronic liver disease is a frequent clinical<br />

presentation <strong>of</strong> ALD, and is associated with a poor outcome. Malnutrition and recent exposure to alcohol<br />

have a negative influence on liver cell regeneration by influencing intracellular proliferative pathways. In<br />

patients with decompensated ALD and biopsy-proven alcoholic ASH, supportive (N-acetylcystein, calories,<br />

vitamins, alcohol abstinence) and anti-inflammatory strategies (corticosteroids) improve short-term survival<br />

but overall prognosis remains poor with a 3-month mortality around 25%. Thus, new strategies to stimulate<br />

liver regeneration and improve function are needed. As liver transplantation is <strong>of</strong>ten not an option at this<br />

stage <strong>of</strong> <strong>the</strong> disease, a minimally invasive regenerative strategy would be welcome.<br />

BONE MARROW CELLS AND LIVER REGENERATION<br />

It has been shown that a subpopulation <strong>of</strong> hepatocytes and cholangiocytes may derive from <strong>the</strong> bone<br />

marrow [1]. Under particular experimental conditions with strong selective pressure (<strong>for</strong> example<br />

immunodeficient animals subject to sublethal liver injury followed by bone marrow transplantation,<br />

fumarylacetatoacetate hydrolase (FAH) deficiency, exposition to hepatotoxin such as retrorsin or carbon<br />

tetrachloride ), hepatocytes <strong>of</strong> bone marrow origin may participate in liver cell repopulation. However, <strong>the</strong><br />

precise mechanisms underlying <strong>the</strong> beneficial effect <strong>of</strong> this promising approach are still under investigation.<br />

Firstly, bone marrow-derived pluripotent cells include hematopoietic and mesenchymal stem cells, as well<br />

as endo<strong>the</strong>lial progenitor cells, and <strong>the</strong> relative contribution <strong>of</strong> each <strong>of</strong> <strong>the</strong>se cellular contingents in liver<br />

repair is ill defined. Secondly, <strong>the</strong>re is much controversy concerning <strong>the</strong> mechanism by which BM-derived<br />

stem cells contribute to liver regeneration (cell fusion, transdifferentiation, local stimulation <strong>of</strong> endogenous<br />

liver cell progenitors, production <strong>of</strong> cytokines and growth factors, remodelling <strong>of</strong> fibrous tissue). Thirdly, <strong>the</strong><br />

mechanisms driving <strong>the</strong>se BM-derived stem cells to <strong>the</strong> site <strong>of</strong> liver injury probably involves multiple factors,<br />

including stromal-derived factor-1 (ie: SDF-1 binding to CXR4 present on HSC surface). Fourthly, <strong>the</strong> rate <strong>of</strong><br />

engraftment <strong>of</strong> <strong>the</strong>se cells in <strong>the</strong> diseased liver is difficult to determine, and seems to vary according to <strong>the</strong><br />

experimental model (< 1% to 50%). Finally, whe<strong>the</strong>r <strong>the</strong> administration <strong>of</strong> G-CSF (to mobilize BM cells and<br />

promote growth factors production), is crucial in governing successful tissue repair is not yet determined.<br />

Regarding safety, <strong>the</strong> small size <strong>of</strong> BM stem cells avoids <strong>the</strong>m to be trapped within <strong>the</strong> sinusoids with <strong>the</strong><br />

potential risk <strong>of</strong> aggregation, ischemia, and increased portal pressure, but <strong>the</strong>re are some concerns about<br />

<strong>the</strong> risk <strong>of</strong> inducing cancer (particularly in culture-expanded cells) and <strong>the</strong> promotion <strong>of</strong> fibrosis (due to <strong>the</strong><br />

presence <strong>of</strong> my<strong>of</strong>ibroblasts among BM cells).<br />

IS BM CELLS MOBILIZATION USING G-CSF ASSOCIATED WITH LIVER REGENERATION<br />

Experimental studies<br />

Animals exposed to various, severe, <strong>of</strong>ten sublethal liver injury (but not related to alcohol) have demonstrated<br />

an improved survival and less severe histological lesions when G-CSF was co-administered [2]. In a<br />

combined toxic liver injury and partial hepatectomy model, G-CSF induced a strong proliferative activity<br />

<strong>of</strong> oval cells (<strong>the</strong> intrahepatic progenitors), as well as <strong>the</strong> generation <strong>of</strong> a subpopulation <strong>of</strong> hepatocytes<br />

originating in <strong>the</strong> bone marrow [3]. Thus, G-CSF seems to improve liver regeneration via both direct (<strong>the</strong><br />

liver stem cell niche) and indirect (contribution to <strong>the</strong> generation <strong>of</strong> bone marrow-derived hepatocytes)<br />

mechanisms. It has also been suggested that G-CSF may contribute to <strong>the</strong> homing <strong>of</strong> bone marrow cells<br />

to <strong>the</strong> injured liver.<br />

Clinical studies<br />

Transposition <strong>of</strong> experimental data with G-CSF into clinical studies with cirrhotics is challenging, with regard<br />

to tolerance (musculoskeletal pain, fever, allergic reactions) and potentially severe side effects (rare cases<br />

<strong>of</strong> splenic rupture). In a small uncontrolled pilot study [4] on advanced cirrhosis (Child-Pugh > 9) <strong>of</strong> mixed<br />

aetiology, subcutaneous administration <strong>of</strong> G-CSF (5 ug/kg twice a day <strong>for</strong> 3 days) was associated with<br />

increased circulating levels <strong>of</strong> CD34+ mobilized BM cells. Tolerance was excellent, although transient<br />

increase in spleen size was reported during treatment. These results were confirmed in a larger group <strong>of</strong><br />

patients with acute decompensated cirrhosis, using ei<strong>the</strong>r a 5 ug/kg/day or a 15 ug/kg/day dose <strong>for</strong> 5 days,<br />

and increased circulating CD34+ cells were associated with a production <strong>of</strong> stromal cell derived factor<br />

(SDF-1), a potent chemo attractant <strong>of</strong> hematopoietic progenitor cells, as well as <strong>the</strong> growth factor VEGF [5].<br />

However, <strong>the</strong>se biological effects didn’t translate into clinical improvement over a 3-week period. To fur<strong>the</strong>r<br />

explore <strong>the</strong> role <strong>of</strong> G-CSF in decompensated cirrhosis, we per<strong>for</strong>med a randomized trial to study <strong>the</strong> shortterm<br />

effect on liver regeneration in patients with decompensated ALD and superimposed ASH (median<br />

Child-Pugh score <strong>of</strong> 10) [6]. Using a 5-day course <strong>of</strong> 10 ug/kg/day G-CSF treatment, circulating CD34+<br />

hematopoietic stem cells were detectable in G-CSF treated patients, but <strong>the</strong> number <strong>of</strong> CD34+ cells was<br />

lower than <strong>the</strong> value obtained in healthy stem cells donors. Using a double immunostaining methods <strong>for</strong> <strong>the</strong><br />

proliferation marker MiB1 and cytokeratin 7 and 18 (immunoreactivity <strong>for</strong> hepatic progenitors and mature<br />

hepatocyte, respectively) per<strong>for</strong>med on baseline and a repeat liver biopsy at day 7, we demonstrated a<br />

significant increase in MiB1+/CK7+ and MiB1+/CK18+ hepatic cells in liver biopsy specimen limited to<br />

G-CSF-treated patients. In addition, changes in circulating CD34+ and MiB1+/CK7+ in liver tissue between<br />

baseline and day-7 values showed a positive correlation (r = 0.65, p < 0.03). Again, <strong>the</strong>se encouraging<br />

results on liver cell proliferation were not associated with any benefits in terms <strong>of</strong> liver function over a<br />

4-week study period.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!