Subscribe to RSS
DOI: 10.1055/s-2002-23204
Toxic Injury to Hepatic Sinusoids: Sinusoidal Obstruction Syndrome (Veno-Occlusive Disease)
Publication History
Publication Date:
27 March 2002 (online)
- ABSTRACT
- HISTORICAL PERSPECTIVES
- CLINICAL FEATURES OF SOS AFTER MYELOABLATIVE THERAPY
- HISTOLOGIC FEATURES OF SOS: HUMAN STUDIES
- PATHOPHYSIOLOGY OF SINUSOIDAL INJURY: ANIMAL MODELS
- PATHOPHYSIOLOGY OF SINUSOIDAL INJURY: CLINICAL STUDIES
- PREVENTION OF SOS IN PATIENTS RECEIVING MYELOABLATIVE THERAPY
- TREATMENT OF PATIENTS WITH SOS FOLLOWING MYELOABLATIVE THERAPY
- ACKNOWLEDGMENTS
- ABBREVIATIONS
- REFERENCES
ABSTRACT
The term veno-occlusive disease of the liver refers to a form of toxic liver injury characterized clinically by the development of hepatomegaly, ascites, and jaundice, and histologically by diffuse damage in the centrilobular zone of the liver. The cardinal histologic features of this injury are marked sinusoidal fibrosis, necrosis of pericentral hepatocytes, and narrowing and eventual fibrosis of central veins. Recent studies suggest that the primary site of the toxic injury is sinusoidal endothelial cells, followed by a series of biologic processes that lead to circulatory compromise of centrilobular hepatocytes, fibrosis, and obstruction of liver blood flow. Thus we propose a more appropriate name for this form of liver injury-sinusoidal obstruction syndrome. This review encompasses historical perspectives, clinical manifestations of sinusoidal obstruction syndrome in the setting of hematopoietic cell transplantation, histologic features of centrilobular injury, and a discussion of the pathophysiology of sinusoidal injury, based on both animal and clinical investigations.
#HISTORICAL PERSPECTIVES
South Africa was the source of the first description of hepatic veno-occlusive disease (VOD) in humans[1] and the first recognition of the vascular origin of the disease.[2] Bras and colleagues in Jamaica coined the name hepatic veno-occlusive disease [3] to describe obliterative fibrosis within small hepatic venules, a feature readily observed by light microscopy. With the recognition that the injury is initiated by changes in the sinusoid[4] (see later section on pathophysiology) and that involvement of hepatic venules is not essential to development of clinical signs and symptoms,[5] we propose that the disease be renamed sinusoidal obstruction syndrome (SOS).
Until the advent of chemotherapy in the 1950s, the only recognized cause of SOS was the ingestion of herbal teas or food sources containing pyrrolizidine alkaloids, found in botanically unrelated plant species (Crotalaria, Heliotropium, Senecio, and, less frequently, Symphytum [comfrey]). Epidemics of SOS have occurred in underdeveloped nations due to consumption of bread made from inadequately winnowed wheat contaminated by seeds from Crotalaria, Heliotropium, and Senecio. [6] [7] There have been numerous reports of SOS related to long-term use of azathioprine for immunosuppression after renal and liver transplantation.[8] [9] [10] [11] [12] [13] Interestingly, there has often been coincident peliosis hepatis, nodular regenerative hyperplasia, or sinusoidal dilatation in the livers of patients with SOS due to azathioprine. Damage to sinusoidal endothelial cells and sometimes to hepatic venular endothelial cells seems to be the common link in these four types of liver injury.[11] [14] [15] [16] [17] [18] Chemotherapeutic agents associated with SOS at conventional doses include gemtuzumab ozogamicin, actinomycin D, dacarbazine, cytosine arabinoside, mithramycin, 6-thioguanine, urethane, indicine N-oxide (a pyrrolizidine alkaloid used as an experimental chemotherapeutic agent to treat leukemia), and chemotherapy combinations.[19] [20] [21] Combinations of irradiation and chemotherapy also may lead to SOS, for example, treatment of nephroblastoma (Wilms' tumor) with dactinomycin and abdominal irradiation.[22] [23] A variant of SOS, radiation-induced liver disease, is seen with radiation doses in excess of 30 to 35 Gy in adults.[24] Although radiation-induced liver disease shares some of the features of SOS, there are significant differences in the clinical presentation, histology, and time course. Recognition of SOS in marrow transplant patients came almost simultaneously from Australia, the United States, and South Africa.[25] [26] [27] Conditioning regimens used for marrow ablation are by far the most common cause of SOS, and will be the focus of this review.
#CLINICAL FEATURES OF SOS AFTER MYELOABLATIVE THERAPY
#Incidence
The incidence of SOS after stem cell transplantation varies from 0 to 50%, due largely to differences in conditioning regimens.[28] [29] [30] [31] [32] [33] There is no liver toxicity from newer, nonmyeloablative conditioning regimens, for example, fludarabine plus low-dose total body irradiation (TBI),[34] but regimens of cyclophosphamide and TBI of greater than 13.2 Gy may cause SOS in half of patients.[28] A major contributor to development of SOS after cyclophosphamide-based conditioning regimens is individual variability in drug metabolism.[35] Overall, the frequency and severity of SOS have fallen dramatically over the past few years, for several reasons: (a) transplantation physicians have retreated from the strategy of dose escalation of conditioning regimens in favor of nonmyeloablative regimens or regimens that do not contain cyclophosphamide; (b) a major risk factor for severe SOS, chronic hepatitis C,[36] has almost disappeared from populations of patients presenting for transplantation; (c) drugs that increase the risk of SOS (e.g., norethisterone[37]) have been removed from transplantation care protocols; and (d) patients are undergoing transplantation sooner after diagnosis of leukemia and arrive for transplantation in a healthier state.
#Clinical Presentation and Diagnosis
Sinusoidal obstruction syndrome is defined by the clinical syndrome of tender hepatomegaly, fluid retention and weight gain, and elevated serum bilirubin that follows cytoreductive therapy, in the absence of other explanations for these signs and symptoms.[28] [31] The onset of SOS is heralded by an increase in liver size, right upper quadrant tenderness, renal sodium retention, and weight gain, occurring 10 to 20 days after the start of cyclophosphamide-based cytoreductive therapy[28] and later after other myeloablative regimens.[33] [38] [39] Patients then develop hyperbilirubinemia, usually before day 20.[28] Some researchers have described a syndrome of ``late VOD'' following conditioning with busulfan-containing regimens, where signs of liver disease are first recognized after day 30.[33] [39]
#Laboratory Studies
Measurement of serum total serum bilirubin is a sensitive test for SOS but not a specific one, because there are many causes of jaundice after transplantation, for example, cholestasis from sepsis, acute graft-versus-host disease (GVHD), hemolysis, renal insufficiency, and cyclosporine therapy.[40] Elevations of serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) can occur in the course of SOS and probably reflect ischemic hepatocyte necrosis, because peak levels are seen weeks after toxin exposure.[41] [42] A serum AST level over 750 U/L in patients with SOS is one marker of a poor prognosis.[41] [42] Several plasma proteins have been reported to be abnormally high in patients with SOS, including endothelial cell markers (hyaluronic acid, von Willebrand factor, plasminogen activator inhibitor-1 [PAI-1], tissue plasminogen activator), thrombopoietin, cytokines (tumor necrosis factor-α[TNF-α], transforming growth factor-β, interleukins-1, -2, -6, and -8, and soluble interleukin-2 [IL-2] receptor), and procollagen peptides.[43] [44] [45] [46] [47] [48] [49] [50] [51] [52] [53] [54] [55] Some laboratory tests are abnormally low in patients with SOS, including the anticoagulant proteins protein C and antithrombin III[56] [57] and platelet counts.[28] [58] It is not clear whether any of these tests have diagnostic or prognostic utility beyond the clinical criteria of weight gain, jaundice, and hepatomegaly, although levels of PAI-1 and IL-8 have been proposed for this role.[53] [55] Serum levels of collagen peptides, however, appear to reflect the extent of sinusoidal fibrosis, probably the most important prognostic variable.[5]
#Ultrasonography, Computed Tomography, and Magnetic Resonance Imaging
Imaging studies of the liver are useful for demonstrating hepatomegaly, ascites, and attenuated hepatic venous flow consistent with SOS,[59] [60] [61] as well as the absence of biliary dilation or infiltrative lesions in the liver and hepatic veins that might also explain hepatomegaly and jaundice.[62] Other ultrasonographic findings that have been reported more frequently in patients with SOS, compared with posttransplantation controls, include gallbladder wall thickening, splenomegaly, visualization of a paraumbilical vein, enlarged portal vein diameter, slow or reversed portal vein flow, high congestion index, portal vein thrombosis, and increased resistive index to hepatic artery flow.[60] [61] [63] [64] [65] [66] [67] Unfortunately, ultrasonographic findings very early in the course of SOS do not appear to add to the information provided by clinical criteria.[59] [68] Later in the course of SOS, especially in patients with severe disease, ultrasonographic evidence of altered liver blood flow, particularly reversal of portal flow and portal vein thrombosis,[66] [67] is more common. There may be value in following vascular parameters as indices of improvement in sinusoidal blood flow.
#Liver Biopsy and Hepatic Venous Pressure Gradient
In cases where the diagnosis is unclear, a transvenous approach that allows both biopsy (using a Mansfield or Cordes forceps or a needle device) and hepatic venous pressure measurements is the most accurate diagnostic test.[69] [70] Percutaneous or laparoscopic needle biopsy are alternative methods of obtaining liver tissue but pose a high risk of bleeding in the thrombocytopenic patient,[71] whereas transvenous biopsy methods can be performed safely with platelet counts as low as 30,000/mm3. In hematopoietic cell transplant patients, a hepatic venous pressure gradient (using an occlusive balloon technique) of greater than 10 mm Hg is highly specific for SOS.[69] [70]
#Differential Diagnosis
Other causes of posttransplantation jaundice seldom lead to renal sodium avidity, rapid weight gain, and hepatomegaly before the onset of jaundice.[40] [72] There are patients who present with jaundice and weight gain that can be confused with SOS.[62] [73] The most common combinations of illnesses that mimic SOS are (a) sepsis syndrome requiring large volumes of crystalloid, followed by renal insufficiency and sepsis-related cholestasis; (b) cholestatic liver disease, hemolysis, and congestive heart failure; and (3) hyperacute GVHD and sepsis syndrome. SOS also may coexist with these disease processes.
#Clinical Course and Prognosis
The severity of SOS has been classified as mild (SOS that is clinically obvious, requires no treatment, and resolves completely), moderate (SOS that causes signs and symptoms requiring treatment such as diuretics or pain medications, but that resolves completely), or severe (SOS that requires treatment but that does not resolve before death or day 100).[28] [31] [32] [74] There is a range of clinical and laboratory findings that correspond to these operational definitions of disease severity (Table [1]).[28] Some patients have subclinical liver damage, evinced by histologic evidence of liver toxicity in the absence of clinical signs and symptoms.[41] Published case fatality rates for SOS after hematopoietic cell transplantation range from 0 to 67%.[28] [30] [31] [32] [33] [75] These figures are dependent on the definition of both SOS and what constitutes fatal SOS. Recovery from SOS was seen in over 70% of patients whose SOS followed cyclophosphamide-containing regimens and in 84% when SOS was caused by other alkylating agents.[28] [32] [38] Despite deep jaundice, patients with severe SOS seldom die of liver failure, but rather from renal and cardiopulmonary failure.[28] [31] [76] [77] A clinically useful model, derived from rates of increase of both bilirubin and weight in the first 2 weeks following transplantation, can predict the outcome of SOS after cyclophosphamide-based regimens.[78] In some patients, there is a bimodal presentation of SOS, that is, clinical signs of SOS appear shortly after transplantation, then wane, then reappear later; this pattern is associated with a worse prognosis.[38] In some cases, signs of SOS resolve, but ascites recurs following development of inflammatory liver disease (e.g., GVHD). A poor prognosis correlates with higher serum AST and ALT values, higher wedged hepatic venous pressure gradient, development of portal vein thrombosis, doubling of the baseline serum creatinine, and decreasing oxygen saturation.[42] [67] [69] [70] [79]
#HISTOLOGIC FEATURES OF SOS: HUMAN STUDIES
Liver biopsy and autopsy specimens over an 80-day period reveal that the histology of injury to sinusoids, hepatocytes, and venules may be rapidly progressive (Fig. [1]). Trichrome connective tissue stains are critical in outlining venules, sinusoids, and hepatocyte cords. Other stains (e.g., Verhoeff-van Gieson and reticulin) are complementary. Serial sections are needed to identify characteristic histologic changes that may not be present in all levels.
#Early Histologic Abnormalities
Histologic data do not elucidate the primary site of injury or molecular events that occur in the sinusoids before clinical signs of SOS appear. The first recognizable histologic change of liver toxicity, occurring 6 to 8 days after cytoreductive or myeloablative therapy, is widening of the subendothelial zone between the basement membrane and the adventitia of central veins and sublobular veins (Fig. [1]B and E).[5] [41] [80] [81] Within the edematous subendothelium are fragmented red cells and bits of cellular debris. Accompanying the venular changes are dilation and engorgement of sinusoids, extravasation of red cells through the space of Disse, and frank necrosis of perivenular hepatocytes-findings that often appear more widespread and severe than the extent of venular injury (Fig. [1]A, B, and E). This is reflected in tissue stained with anticytokeratin antibody (a marker of hepatocyte viability) that shows markedly diminished staining of centrizonal hepatocytes.[81] One morphologic consequence of sinusoidal obstruction, ischemia, elevated sinusoidal pressures, and fragmentation of hepatocyte cords is dislodgement of clusters of hepatocytes that may flow retrograde into portal veins or embolize through disrupted pores into lumina of damaged central veins (Fig. [1]C). Immunohistology studies demonstrate deposition of fibrinogen and Factor VIII/von Willebrand factor, but not platelet antigens, in the perivenular zone and in the widened subendothelial space of venules (but not within venular lumens), corresponding to clogging of pores that drain sinusoids into the venules (Fig. [1]D).[81] Electron microscopy studies show closure of fenestrae in sinusoidal endothelial cells and accumulation of extracellular material (collagen, possibly fibrin) in sinusoidal pores.[80]
#Later Histologic Finding: Stellate Cell Proliferation and Collagenization
Within 2 weeks of the onset of clinical signs of SOS, curvilinear deposits of extracellular matrix can be seen in subendothelial spaces and in sinusoids (Fig. [1]C and E). Immunostaining for activated stellate cells with α smooth muscle actin antibodies demonstrates a marked increase in the number of stellate cells lining the sinusoids (Fig. [1]G)[82] [83] that are clogged by types I, III, and IV collagen.[81] We speculate that the bimodal pattern of clinical manifestations of SOS that is seen in some patients reflects early sinusoidal injury followed later by proliferation of stellate cells and deposition of matrix in sinusoids. The later stages of fatal SOS (i.e., beyond day 50 following cyclophosphamide-based regimens or earlier, following gemtuzumab ozogamicin infusions[19] [20]) are characterized by extensive collagenization of sinusoids and venules (Fig. [1]F and H). In some cases, there is coalescence of extinguished perivenular zones with fibrous bridging between central veins, simulating cardiac cirrhosis (Fig. [1]H).
#Correlation of Histologic Findings with Clinical Signs
In retrospective autopsy studies, 20 to 30% of cases with occluded venules were without clinical symptoms.[31] [41] Furthermore, several perivenular lesions were reported to correlate with signs of SOS in the absence of venular occlusion.[41] A coded review of histologic features in a cohort of 76 consecutive necropsy patients found that the strongest statistical associations with clinically severe SOS were hepatocyte necrosis, sinusoidal fibrosis, eccentric localized thickening of the subendothelial zone of the venule, phlebosclerosis, and an estimate of venular narrowing.[5] Moreover, the number of such histologic changes present was proportional to the clinical severity of SOS.
#PATHOPHYSIOLOGY OF SINUSOIDAL INJURY: ANIMAL MODELS
In vitro studies have shown that sinusoidal endothelial cells are more susceptible than hepatocytes to drugs that cause SOS.[17] [84] [85] This is consistent with in vivo studies conducted in the rat monocrotaline model. Monocrotaline, the pyrrolizidine alkaloid found in Crotalaria, is one of the best-studied toxins involved in SOS. The monocrotaline model of SOS has the same histologic characteristics as the human disease, as well as the same ``clinical features,'' i.e., hyperbilirubinemia, hepatomegaly, and ascites formation. In this model the first morphologic change noted by electron microscopy is loss of sinusoidal endothelial cell fenestration and the appearance of gaps in the sinusoidal endothelial cell barrier.[4] Studies with in vivo microscopy and confirmation by electron microscopy have shown that sinusoidal endothelial cells round up, and red blood cells begin to penetrate into the space of Disse beneath the rounded up endothelial cells and dissect off the sinusoidal lining. The sloughed sinusoidal lining cells (i.e., Kupffer cells, sinusoidal endothelial cells, and stellate cells) embolize downstream and obstruct sinusoidal flow.[86] By the time hepatocyte necrosis is observed, there is extensive denudation of the sinusoidal lining. Early on there is loss of Kupffer cells, but there is a significant influx of monocytes within the sinusoids, which exacerbates the obstruction of sinusoidal flow by the embolized sinusoidal lining cells. In summary, the major findings of these studies are that the initiating event of SOS is rounding up or swelling of sinusoidal endothelial cells and that this leads to dissection of the sinusoidal lining, which embolizes and blocks the microcirculation.
In addition to the morphologic changes described above, a number of biochemical changes have been observed in the experimental studies. Drugs and toxins that cause SOS profoundly deplete sinusoidal endothelial cell glutathione prior to cell death, and support of sinusoidal endothelial cell glutathione prevents cell death.[84] [85] [87] Continuous infusion of glutathione or N-acetylcysteine into the portal vein prevents the morphologic changes of SOS observed by light microscopy, electron microscopy, and in vivo microscopy, as well as the ``clinical features'' of SOS.[88] Infusion of glutathione 24 hours after treatment with monocrotaline attenuated, but did not prevent, SOS. Interestingly, discontinuation of the glutathione infusion several days after monocrotaline has been eliminated from the rat leads to accelerated development of full-blown SOS. Thus, initially glutathione may protect (partially) by preventing profound glutathione depletion. However another mechanism for protection must be invoked to explain glutathione protection several days after monocrotaline has been eliminated. Because full-blown SOS in this model normally takes 72 hours to develop, the development of SOS within 24 hours after discontinuation of glutathione indicates that glutathione is suppressing a persistent change in the sinusoid. The benefit of GSH described above is consistent with improved survival after administration of monocrotaline in rats treated with glutathione monoethyl ester.[89]
One possible explanation for the rounding up of sinusoidal endothelial cells may be increased activity of matrix metalloproteinases (MMPs). Because MMPs degrade extracellular matrix, increased MMP activity on the ablumenal side of the sinusoidal endothelial cell would allow the cells to let loose from the space of Disse. In the experimental model, de novo synthesis of MMP-9 (gelatinase B) and increased MMP-9 activity occur 12 hours after monocrotaline, which coincides with rounding up of the sinusoidal endothelial cells.[90] Inhibition of MMP activity completely prevents SOS. MMP expression and activity are regulated by redox status and can be suppressed by glutathione and N-acetylcysteine.[91] [92] [93] [94] Thus, the protective effect of glutathione and N-acetylcysteine may be (partially) due to inhibition of MMP activity.
In the in vivo model, hepatic vein nitric oxide decreases in parallel with the changes in sinusoidal flow.[95] Inhibition of nitric oxide elicits severe SOS due to a subtoxic dose of monocrotaline, whereas nitric oxide (NO) precursors prevent the morphologic changes and ``clinical features'' of SOS.[95] Interestingly, tonic release of NO by endothelial cells reduces MMP-9 expression, whereas inhibition of NO synthesis increases cytokine-stimulated MMP-9 expression.[96] Future studies will need to determine whether the contribution of decreased hepatic NO to SOS occurs through increased MMP-9 synthesis.
Although SOS is defined as a nonthrombotic obstruction of flow, the issue of clotting has been a recurring topic, based largely on plasma studies in patients with SOS.[97] However, electron microscopy of pyrrolizidine alkaloid-induced SOS in humans did not detect clotting.[98] Sequential observations during the development of SOS in the experimental model by in vivo microscopy and electron microscopy have not demonstrated any evidence of clotting.[4]
#PATHOPHYSIOLOGY OF SINUSOIDAL INJURY: CLINICAL STUDIES
Pharmacokinetics of chemotherapy drugs and radiation dosimetry are the most important determinants of SOS after transplantation, but there are also events that follow toxic injury that appear to be prognostically important. In addition, some patients are at risk because of risk factors for fatal SOS, for example, hepatitis C, nonalcoholic steatohepatitis, and systemic bacterial or viral infection before the start of cytoreductive therapy.[28] [36] Previous radiation therapy that involved the liver and previous stem cell transplantation are also predisposing factors.[28] [37] [38] [99] Two observations about SOS in patients provide clues as to the mechanism of disease. First, unlike other intrinsic liver diseases, the signs and symptoms of portal hypertension precede evidence of parenchymal damage. In SOS, disruption of the liver circulation is the cause and not the consequence of the parenchymal disease. Second, involvement of the hepatic veins is not essential to the development of clinical signs: 45% of patients with mild or moderate disease and 25% of patients with severe SOS did not have occluded hepatic venules at autopsy.[5] Occlusion of central veins is associated with more severe disease and the development of ascites.[5] [100] This suggests that venular lesions exacerbate the circulatory impairment that occurs at the level of the sinusoid.
#Chemotherapy Drugs
Cyclophosphamide is common to the transplantation conditioning regimens with the highest incidence of fatal SOS.[28] The metabolism of cyclophosphamide is highly variable; patients who generate a greater quantity of toxic metabolites are more likely to develop severe SOS following conditioning with cyclophosphamide and TBI.[35] [101] The liver toxin generated by cyclophosphamide metabolism is acrolein (a metabolite formed simultaneously along with the desired metabolite, phosphoramide mustard), via mechanisms dependent on glutathione.[84] [89] [102] [103] Busulfan is another component of regimens with a high frequency of SOS. A relationship between busulfan exposure (measured by the area under the curve or average steady-state concentration) following oral dosing and the toxicity of conditioning therapy has been reported.[104] [105] [106] [107] [108] However, in adults with chronic myeloid leukemia in chronic phase and children with acute leukemia, there is no relationship between busulfan exposure and SOS.[106] [109] [110] [111] Busulfan may contribute to liver injury by inducing oxidative stress, reducing glutathione levels in hepatocytes and sinusoidal endothelial cells,[112] and altering cyclophosphamide metabolism.[101] When cyclophosphamide is given before busulfan, a lower frequency of SOS results, suggesting that busulfan predisposes patients to cyclophosphamide toxicity.[113] Most transplantation centers now dose oral busulfan according to its metabolism or give intravenous doses of busulfan, with more predictable kinetics and less overall toxicity.[114] [115] SOS also has been described as dose limiting for regimens that contain high dose BCNU (1,3-bis(2-chbroethyl)-1-nitrosourea), carboplatin, or cytarabine.[75]
#Total Body Irradiation
The doses of TBI given in the setting of hematopoietic cell transplantation are in the range of 10 to 16 Gy, far less that the dose of liver irradiation that causes radiation-induced liver disease.[24] [116] In combination with cyclophosphamide, however, there is a clear relationship between the total dose of TBI and the frequency of severe SOS.[28] [35] The relationship of irradiation technique to SOS is controversial, with one study showing such a relationship[117] and three studies showing no association.[28] [118] [119] The source of radiation for TBI also may be important, because the dose rate differs between radiation from opposing cobalt sources versus a linear accelerator.
#Intrahepatic Coagulation
Some see SOS as a disease of disordered coagulation, in which damage to endothelium in the sinusoids and central veins leads to exposure of tissue factor and initiation of the coagulation cascade, a process abetted by low circulating levels of the natural anticoagulants protein C and antithrombin III, elaboration of cytokines that stimulate coagulation, high plasma levels of tissue plasminogen activator, and reduced fibrinolysis (reflected by elevated plasma levels of PAI-1).[53] [57] [120] [121] [122] [123] [124] However, heparin and antithrombin III infusions are ineffective in preventing SOS, and thrombolytic therapy effects improvement in only a minority of patients.[32] [79] [125] [126] [127] [128] [129] [130] [131] Genetic disorders predisposing to coagulation (Factor V Leiden and prothrombin gene 20210 G-A) have weak or no associations with SOS after transplantation.[132] [133] Disordered coagulation in SOS is probably an epiphenomenon secondary to widespread centrilobular damage, not a cause of damage.
#Stellate Cells and Sinusoidal Fibrosis
Several series have documented the early appearance of a procollagen peptide in serum in patients who develop more severe SOS, along with inhibitors of fibrolysis,[47] [48] [52] [134] consistent with the intense fibrosis in centrilobular sinusoids and venular walls that is common in fatal SOS (Fig. [1]F and H).[5] [19] [20] The stimuli for stellate cell activation and proliferation in the transplantation setting have not been identified; candidates include centrilobular hypoxia, endotoxemia, Kupffer cell damage, loss of sinusoidal endothelial cells, and a melange of circulating growth factors and cytokines (platelet-derived growth factor, transforming growth factor-β, TNF-α, IL-8, and soluble IL-2 receptor).[44] [45] [54] [55] [135] [136] [137] Not all series have found cytokine levels to be elevated in patients with SOS.[50] [138]
#Vasoactive Mediators
Serum levels of endothelin-1 are elevated in patients with SOS.[138] [139] Low plasma levels of nitrate are also seen during conditioning therapy.[138] [139] These data are consistent with the hypothesis that endothelin-1 is a mediator of hepatic sinusoidal constriction, unopposed by a vasorelaxant effect of endothelial nitric oxide.
#Renal Pathophysiology in SOS
Decreasing fractional excretion of sodium occurs just before the clinical signs of SOS become apparent, reflecting both sinusoidal hypertension[69] [70] and renal tubular injury caused by metabolites of cyclophosphamide.[140] Plasma levels of nitrate increase sharply during fluid accumulation in patients who develop moderate to severe SOS.[139]
#PREVENTION OF SOS IN PATIENTS RECEIVING MYELOABLATIVE THERAPY
The only certain way to prevent fatal SOS is to avoid giving hepatotoxic conditioning therapy to patients at high risk: (a) nonfatal SOS following previous chemotherapy or radiation therapy to the liver; (b) second transplants with myeloablative regimens[99]; (c) hepatic inflammation[28] [36]; and (d) extensive hepatic fibrosis or cirrhosis.[141] [142]
#Alterations of Doses of Conditioning Therapy
The use of nonmyeloablative regimens that contain no liver toxins, followed by allogeneic transplantation, may be the only method of avoiding SOS in patients with underlying hepatic fibrosis.[34] Altering myeloablative conditioning regimens has been reported to lessen the risk of SOS, for example, adjusting oral doses of busulfan in individual patients based on busulfan disposition[104] [143] or giving busulfan as an intravenous preparation.[115] However, other studies of busulfan dosing based on steady-state concentrations have not shown benefit in preventing SOS.[109] [110] [111] [144] Giving intravenous busulfan along with cyclophosphamide does not eliminate fatal SOS, lending credence to cyclophosphamide as the more important liver toxin in this regimen.[115] Administering cyclophosphamide before busulfan[113] and giving irradiation doses of 10 Gy at 2 to 4 cGy/min may result in a lower incidence of SOS.[32] [145] [146] Fractionated doses of BCNU are associated with a lower incidence of SOS than are single doses.[147] Shielding the liver during TBI will lessen liver injury but leads to relapse of underlying hematologic disease.[148] Another approach for conditioning a patient who has risk factors for fatal SOS is to choose a regimen that does not contain cyclophosphamide.
#Anticoagulant or Antithrombin Infusions
Anticoagulation with heparin can be given safely if the partial thromboplastin time is monitored carefully.[149] [150] Two randomized studies have reported a reduction in nonfatal SOS with heparin, but had little power to show a benefit in preventing severe SOS.[150] [151] Four additional studies of anticoagulation could find no benefit.[32] [146] [149] [152] Nonetheless, some centers routinely use heparin infusions or low-molecular-weight heparin subcutaneously.[153] [154] Prophylactic antithrombin III infusions effect no reduction in the frequency or severity of SOS despite restoring plasma antithrombin III levels to normal.[125]
#Other Medical Prophylaxis
Ursodeoxycholic acid (ursodiol) has been reported to reduce the severity of posttransplantation SOS in two randomized trials,[155] [156] but was without effect in a large randomized Scandinavian study.[157] Two trials have administered prostaglandin E1 by continuous infusion with no evidence that the frequency of fatal SOS was affected.[158] [159] Because some studies have reported elevated levels of cytokines (including TNF-α) in association with SOS,[44] anticytokine strategies have been studied. Several placebo-controlled, randomized trials have shown pentoxifylline, an inhibitor of TNF-α release, to be ineffective in preventing SOS.[160] [161]
#TREATMENT OF PATIENTS WITH SOS FOLLOWING MYELOABLATIVE THERAPY
Because 70 to 85% of patients recover spontaneously, treatment of SOS involves management of sodium and water balance with diuretics and repeated paracenteses for ascites that is associated with discomfort or pulmonary compromise.[28] Renal and pulmonary failure in patients with severe SOS are managed with hemodialysis and mechanical ventilation, albeit with little impact on outcome.[28] [76] [77] [162] In severely ill hematopoietic stem cell transplant patients with SOS and organ failure, there are guidelines that have defined futility of treatment, allowing patients and their physicians to eschew life-support measures when appropriate, or to consider liver transplantation in special circumstances.[162] There is no completely satisfactory treatment for severe SOS, but the literature describes the following approaches.
#Thrombolytic Therapy
Tissue plasminogen activator and heparin infusions show evidence of efficacy in less than a third of patients with severe SOS.[79] [126] [127] [128] [129] [130] [131] [163] In one large series, there were no responses when patients had either renal or pulmonary failure.[79] Thrombolytic therapy is further limited by the risk of fatal intracerebral and pulmonary bleeding.[79] [131] [164] [165] Thrombolytic therapy might be considered for treatment of patients with SOS whose prognostic indices[78] point to a greater than 20% estimated risk of fatality provided that renal and lung function are intact.
#Defibrotide
Defibrotide, a single-stranded polydeoxyribonucleotide drug derived from animal tissue, has antithrombotic, antiischemic, and thrombolytic properties.[166] Uncontrolled trials with defibrotide in over 100 patients with moderate to severe SOS showed complete resolution in 35 to 55% of patients and no evidence of serious toxicity.[167] [168] Recovery of patients with severe SOS and multiorgan failure is unusual, lending credence to reports of complete recovery following defibrotide infusion.
#Other Medical Therapies
Three patients with clinical evidence of moderate to severe SOS have been treated with intravenous N-acetylcysteine at 50 to 150 mg/kg/day for 2 to 4 weeks, during which serum bilirubin and inflammatory markers improved.[137] Patients with SOS have been treated with infusions of human antithrombin III concentrate or activated protein C[125] [169] [170] without clear evidence of efficacy. There are also anecdotal reports of improvement after therapy with prostaglandin E1,[171] prednisone,[172] topical nitrate,[173] and vitamin E/glutamine therapy.[174]
#Transjugular Intrahepatic Portosystemic Shunt
Transhepatic intrahepatic portosystemic shunts (TIPS) have been placed in patients with SOS after hematopoietic cell transplantation.[175] [176] [177] [178] TIPS placement reduces portal pressure and appears effective in mobilizing ascites, but does not have an effect on either serum bilirubin levels or patient outcomes.[177] [178] [179] In one case, TIPS led to acute respiratory distress syndrome that was fatal.[180]
#Surgical Approaches
Two patients with SOS underwent successful portosystemic shunts for persistent ascites, but liver dysfunction had resolved long before these shunts were placed.[181] [182] Peritoneovenous shunts for intractable ascites have been unsuccessful. Eleven patients have received liver transplants for severe SOS,[165] [183] [184] [185] [186] [187] [188] [189] four of whom were midterm survivors.[183] [186] [188] [189] When severe SOS develops in a patient with a benign condition (a rare event) or in a patient with a favorable outcome posttransplantation (e.g., chronic myelogenous leukemia in chronic phase), liver transplantation should be considered.
#ACKNOWLEDGMENTS
Dr. DeLeve is supported by the National Institutes of Health, NIDDK, Grant DK46357; Drs. McDonald and Shulman are supported by the National Institutes of Health, National Cancer Institute, Grants CA 15704 and CA 18029.
#ABBREVIATIONS
ALT alanine aminotransferase
AST aspartate aminotransferase
GSH reduced glutathione
GVHD graft-versus-host disease
MMP matrix metalloproteinase
PAI-1 plasminogen activator inhibitor-1
SOS sinusoidal obstruction syndrome
TBI total body irradiation
TIPS transjugular intrahepatic portosystemic shunt
VOD veno-occlusive disease of the liver

Figure 1 Photomicrographs illustrating the evolution of histologic abnormalities of SOS. (A and B) Fatal SOS 23 days postautograft (hematoxylin and eosin stain). (A) Hemorrhagic necrosis of zone 3 with sparing of zone 1 (PV, portal vein). (B) High-power methacrylate thin section showing early changes of concentric venular luminal narrowing by a widened subendothelial zone (double arrow) containing entrapped red cells. The surrounding changes include anuclear clusters of necrotic hepatocytes, disrupted sinusoids, and hemorrhage into the space of Disse. (C) Severe SOS 21 days postallograft (Gomori trichrome stain). Hepatocyte dropout, sinusoidal hemorrhage, and embolization of a cluster of hepatocytes (arrow) into a partially collagenized venule. (D) Immunostained liver section of early SOS with anti-Factor VIII/von Willebrand factor, showing intense perivenular and adventitial staining, loss of the hepatocyte background staining in zone 3, and preservation of portal and zone 1 anatomy. A similar perivenular pattern of deposition was seen with immunofluorescent anti-fibrogen immunostaining, whereas anti-platelet antigen immunostaining was negative.[81] (E) Fatal SOS 31 days postallograft (Mallory trichrome stain). A small sublobular venule is nearly occluded by loose extracellular matrix and red cells within the widened subendothelial zone (double arrows). Extensive necrosis has obliterated the liver cords, which are replaced by fine curvilinear strands of connective tissue admixed with red cells. (F and G) Fatal sinusoidal obstruction syndrome 37 days after (gemtuzumab ozogamicin infusion for relapsed acute myelogenous leukemia posttransplantation. (F) Extensive zone 3 sinusoidal fibrosis adjacent to a nonoccluded sublobular venule (Mallory trichrome stain). (G) Immunostain of the same specimen with anti-α smooth muscle actin showing proliferation of activated hepatic stellate cells lining perivenular sinusoids. (H) Fatal SOS 67 days postallograft (Mallory trichrome stain), showing a pattern of ``reverse'' cirrhosis with confluent bridging between collapsed, fibrotic perivenular zones containing venules occluded by fibrous matrix.

REFERENCES
- 1 Willmot F C, Robertson G W. Senecio disease, or cirrhosis of the liver due to senecio poisoning. Lancet . 1920; 2 848-849
- 2 Selzer G, Parker R GF. Senecio poisoning exhibiting as Chiari's syndrome: a report on twelve cases. Am J Pathol . 1950; 27 885-887
- 3 Bras G, Jeliffe D B, Stuart K L. Veno-occlusive disease of the liver with non-portal type of cirrhosis occurring in Jamaica. Arch Pathol . 1954; 57 285-300
- 4 DeLeve L D, McCuskey R S, Wang X. Characterization of a reproducible rat model of hepatic veno-occlusive disease. Hepatology . 1999; 29 1779-1791
- 5 Shulman H M, Fisher L B, Schoch H G. Venocclusive disease of the liver after marrow transplantation: histologic correlates of clinical signs and symptoms. Hepatology . 1994; 19 1171-1180
- 6 Mohabbat O, Younos M S, Merzad A A. An outbreak of hepatic veno-occlusive disease in north-western Afghanistan. Lancet . 1976; 2 269-271
- 7 Tandon H D, Tandon B N, Mattocks A R. An epidemic of veno-occlusive disease of the liver in Afghanistan. Pathologic features. Am J Gastroenterol . 1978; 70 607-613
- 8 Eisenhauer T, Hartmann H, Rumpf K W. Favourable outcome of hepatic veno-occlusive disease in a renal transplant patient receiving azathioprine, treated by portacaval shunt. Report of a case and review of the literature. Digestion . 1984; 30 185-190
- 9 Katzka D A, Saul S H, Jorkasky D. Azathioprine and hepatic venocclusive disease in renal transplant patients. Gastroenterology . 1986; 90 446-454
- 10 Read A E, Weisner R H, LaBrecque D R. Hepatic venocclusive disease associated with renal transplantation and azathioprine therapy. Ann Intern Med . 1986; 104 651-655
- 11 Haboubi N Y, Ali H H, Whitwell H L. Role of endothelial cell injury in the spectrum of azathioprine-induced liver disease after renal transplant: light microscopy and ultrastructural observations. Am J Gastroenterol . 1988; 83 256-261
- 12 Liano F, Moreno A, Matesanz R. Veno-occlusive hepatic disease of the liver in renal transplantation: is azathioprine the cause?. Nephron . 1989; 51 509-516
- 13 Sterneck M, Wiesner R, Ascher N. Azathioprine hepatotoxicity after liver transplantation [Review]. Hepatology . 1991; 14 806-810
- 14 de Araujo S M, Gerard F, Chossegros P. Vascular hepatotoxicity related to heroin addiction. Virchows Arch [A] . 1990; 417 497-503
- 15 Trigueiro de Araujo S M, Gerard F, Chossegros P. Lack of hepatocyte involvement in the genesis of the sinusoidal dilatation related to heroin addiction: a morphometric study. Virchows Arch [A] . 1992; 420 149-153
- 16 Zafrani E S, Cazier A, Baudelot A M. Ultrastructural lesions of the liver in human peliosis. A report of 12 cases. Am J Pathol . 1984; 114 349-359
- 17 DeLeve L D, Wang X, Kuhlenkamp J F. Toxicity of azathioprine and monocrotaline in murine sinusoidal endothelial cells and hepatocytes: the role of glutathione and relevance to hepatic veno-occlusive disease. Hepatology . 1996; 23 589-599
- 18 DeLeve L D. Glutathione defense in non-parenchymal cells [Review]. Semin Liver Dis . 1998; 18 403-413
- 19 Giles F J, Kantarjian H M, Kornblau S M. Mylotarg (gemtuzumab ozogamicin) therapy is associated with hepatic venoocclusive disease in patients who have not received stem cell transplantation. Cancer . 2001; 92 406-413
- 20 Rajvanshi P, Shulman H M, Sievers E L. Hepatic sinusoidal obstruction following Gemtuzumab ozogamicin (Mylotarg). Blood (in press). 2002;
- 21 King P D, Perry M C. Hepatotoxicity of chemotherapy [Review]. Oncologist . 2001; 6 162-176
- 22 Tornesello A, Piciacchia D, Mastrangelo S. Veno-occlusive disease of the liver in right-sided Wilms' tumours [Review]. Eur J Cancer . 1998; 34 1220-1223
- 23 Czauderna P, Katski K, Kowalczyk J. Venoocclusive liver disease (VOD) as a complication of Wilms' tumour management in the series of consecutive 206 patients. Eur J Pediatr Surg . 2000; 10 300-303
- 24 Lawrence T S, Robertson J M, Anscher M S. Hepatic toxicity resulting from cancer treatment [Review]. Int J Radiat Oncol Biol Phys . 1995; 31 1237-1248
- 25 Vowels M, Lam-Po-Tang R, Zagars G. Total body irradiation and Budd-Chiari syndrome. Pathology . 1979; 11 306
- 26 Berk P D, Popper H, Krueger G RF. Veno-occlusive disease of the liver after allogeneic bone marrow transplantation. Ann Intern Med . 1979; 90 158-164
- 27 Jacobs P, Miller J L, Uys C J. Fatal veno-occlusive disease of the liver after chemotherapy, whole-body irradiation and bone marrow transplantation for refractory acute leukaemia. S Afr Med J . 1979; 55 5-10
- 28 McDonald G B, Hinds M S, Fisher L B. Venocclusive disease of the liver and multiorgan failure after bone marrow transplantation: a cohort study of 355 patients. Ann Intern Med . 1993; 118 255-267
- 29 McDonald G B, Sharma P, Matthews D E. Venocclusive disease of the liver after bone marrow transplantation: diagnosis, incidence, and predisposing factors. Hepatology . 1984; 4 116-122
- 30 Ganem G, Saint-Marc Girardin F M, Kuentz M. Venocclusive disease of the liver after allogeneic bone marrow transplantation in man. Int J Radiat Oncol Biol Phys . 1988; 14 879-884
- 31 Jones R J, Lee K S, Beschorner W E. Veno-occlusive disease of the liver following bone marrow transplantation. Transplantation . 1987; 44 778-783
- 32 Carreras E, Bertz H, Arcese W. Incidence and outcome of hepatic veno-occlusive disease after blood or marrow transplantation: a prospective cohort study of the European group for blood and marrow transplantation. Blood . 1998; 92 3599-3604
- 33 Hasegawa S, Horibe K, Kawabe T. Veno-occlusive disease of the liver after allogeneic bone marrow transplantation in children with hematologic malignancies: incidence, onset time and risk factors. Bone Marrow Transplant . 1998; 22 1191-1197
- 34 McSweeney P A, Niederwieser D, Shizuru J A. Hematopoietic cell transplantation in older patients with hematologic malignancies: replacing high-dose cytotoxic therapy with graft-versus-tumor effects. Blood . 2001; 97 3390-3400
- 35 McDonald G B, Ren S, Bouvier M E. Venocclusive disease of the liver and cyclophosphamide pharmacokinetics: a prospective study in marrow transplant patients [Abstract]. Hepatology . 1999; 30 314
- 36 Strasser S I, Myerson D, Spurgeon C L. Hepatitis C virus infection after bone marrow transplantation: a cohort study with 10 year follow-up. Hepatology . 1999; 29 1893-1899
- 37 Hagglund H, Remberger M, Klaesson S. Norethisterone treatment, a major risk factor for veno-occlusive disease in the liver after allogeneic bone marrow transplantation. Blood . 1998; 92 4568-4572
- 38 Lee J L, Gooley T, Bensinger W. Venocclusive disease of the liver after busulfan, melphalan, and thioTEPA conditioning therapy: incidence, risk factors, and outcome. Biol Blood Marrow Transplantation . 1999; 5 306-315
- 39 Toh H C, McAfee S L, Sackstein R. Late onset veno-occlusive disease following high-dose chemotherapy and stem cell transplantation. Bone Marrow Transplant . 1999; 24 891-895
- 40 Strasser S I, McDonald G B. Hepatobiliary complications of hematopoietic stem cell transplantation. In: Schiff ER, Sorrell MF, Maddrey WC, eds. Schiff's Diseases of the Liver, 8th ed Philadelphia: Lippincott-Raven, 1999: 1617-1641
- 41 Shulman H M, McDonald G B, Matthews D. An analysis of hepatic venocclusive disease and centrilobular hepatic degeneration following bone marrow transplantation. Gastroenterology . 1980; 79 1178-1191
- 42 Strasser S I, McDonald S J, Schoch H G. Severe hepatocellular injury after hematopoietic cell transplant: incidence and etiology in 2136 consecutive patients [Abstract]. Hepatology . 2000; 32 299
- 43 Blann A, Collins P. Von Willebrand factor, angiotensin converting enzyme and endothelial cell damage in veno-occlusive disease and bone marrow transplantation. Thromb Res . 1992; 66 617-618
- 44 Holler E, Kolb H J, Moller A. Increased serum levels of tumor necrosis factor-a precede major complications of bone marrow transplantation. Blood . 1990; 75 1011-1016
- 45 Anscher M S, Peters W P, Reisenbichler H. Transforming growth factor βas a predictor of liver and lung fibrosis after autologous bone marrow transplantation for advanced breast cancer. N Engl J Med . 1993; 328 1592-1598
- 46 Oh H, Tahara T, Bouvier M. Plasma thrombopoietin levels in marrow transplant patients with veno-occlusive disease of the liver. Bone Marrow Transplant . 1998; 22 675-679
- 47 Eltumi M, Trivedi P, Hobbs J. Monitoring of veno-occlusive disease after bone marrow transplantation by serum aminopropeptide of type III procollagen. Lancet . 1993; 342 518-521
- 48 Rio B, Bauduer F, Arrago J P. N-terminal peptide of type III procollagen: a marker for the development of hepatic veno-occlusive disease after BMT and a basis for determining the timing of prophylactic heparin. Bone Marrow Transplant . 1993; 11 471-472
- 49 Park Y D, Yasui M, Yoshimoto T. Changes in hemostatic parameters in hepatic veno-occlusive disease following bone marrow transplantation. Bone Marrow Transplant . 1997; 19 915-920
- 50 Murase T, Jirtle R L, McDonald G B. Transforming growth factor-beta plasma concentrations in patients with leukemia and lymphoma receiving chemoradiotherapy and marrow transplantation [Letter]. Blood . 1994; 83 2383
- 51 Farrand A L, Bouvier M E, McDonald G B. Plasma levels of the propeptide of type III collagen predict the severity of venocclusive disease of the liver [Abstract]. Hepatology . 1996; 24 508
- 52 Heikinheimo M, Halila R, Fasth A. Serum procollagen type III is an early and sensitive marker for venocclusive disease of the liver in children undergoing bone marrow transplantation. Blood . 1994; 83 3036-3040
- 53 Salat C, Holler E, Kolb H J. Plasminogen activator inhibitor-1 confirms the diagnosis of hepatic veno-occlusive disease in patients with hyperbilirubinemia after bone marrow transplantation. Blood . 1997; 89 2184-2188
- 54 Remberger M, Ringden O. Increased levels of soluble interleukin-2 receptor in veno-occlusive disease of the liver after allogenic bone marrow transplantation. Transplantation . 1995; 60 1293-1299
- 55 Remberger M, Ringden O. Serum levels of cytokines after bone marrow transplantation: increased IL-8 levels during severe veno-occlusive disease of the liver. Eur J Haematol . 1997; 59 254-262
- 56 Scrobohaci M L, Drouet L, Monem-Mansi A. Liver venocclusive disease after bone marrow transplantation. Changes in coagulation parameters and endothelial markers. Thromb Res . 1991; 63 509-519
- 57 Faioni E M, Krachmalnicoff A, Bearman S I. Naturally occurring anticoagulants and bone marrow transplantation: plasma protein C predicts the development of venocclusive disease of the liver. Blood . 1993; 81 3458-3462
- 58 Rio B, Andreu G, Nicod A. Thrombocytopenia in venocclusive disease after bone marrow transplantation or chemotherapy. Blood . 1986; 67 1773-1776
- 59 Hommeyer S C, Teffey S A, Jacobson A F. Venocclusive disease of the liver: prospective study of US evaluation. Radiology . 1992; 184 683-686
- 60 Herbetko J, Grigg A P, Buckley A R. Veno-occlusive liver disease after bone marrow transplantation: findings at duplex sonography. AJR . 1992; 158 1001-1005
- 61 Lassau N, Leclere J, Auperin A. Hepatic veno-occlusive disease after myeloablative treatment and bone marrow transplantation: value of gray-scale and Doppler US in 100 patients. Radiology . 1997; 204 545-552
- 62 Costa F, Choy C G, Seiter K. Hepatic outflow obstruction and liver failure due to leukemic cell infiltration in chronic lymphocytic leukemia. Leuk Lymphoma . 1998; 30 403-410
- 63 Brown B P, Abu-Yousef M, Farner R. Doppler sonography: a noninvasive method for evaluation of hepatic venocclusive disease. AJR . 1990; 154 721-724
- 64 Deeg K H, Glockel U, Richter R. Diagnosis of veno-occlusive disease of the liver by color-coded Doppler sonography. Pediatr Radiol . 1993; 23 134-136
- 65 Nicolau C, Bru C, Carreras E. Sonographic diagnosis and hemodynamic correlation in veno-occlusive disease of the liver. J Ultrasound Med . 1993; 12 437-440
- 66 Grigg A, Gibson R, Bardy P. Acute portal vein thrombosis after autologous stem cell transplantation. Bone Marrow Transplant . 1996; 18 949-953
- 67 Kikuchi K, Rudolph R, Murakami C. Portal vein thrombosis after hematopoietic cell transplantation: frequency, treatment, and outcome. Bone Marrow Transplantation (in press). 2002;
- 68 McCarville M B, Hoffer F A, Howard S C. Hepatic veno-occlusive disease in children undergoing bone-marrow transplantation: usefulness of sonographic findings. Pediatr Radiol . 2001; 31 102-105
- 69 Carreras E, Granena A, Navasa M. Transjugular liver biopsy in bone marrow transplantation. Bone Marrow Transplant . 1993; 11 21-26
- 70 Shulman H M, Gooley T, Dudley M D. Utility of transvenous liver biopsies and wedged hepatic venous pressure measurements in sixty marrow transplant recipients. Transplantation . 1995; 59 1015-1022
- 71 Sharma P, McDonald G B, Banaji M. The risk of bleeding after percutaneous liver biopsy: relation to platelet count. J Clin Gastroenterol . 1982; 4 451-453
- 72 Zimmerman H J, Fang M, Utili R. Jaundice due to bacterial infection. Gastroenterology . 1979; 28 249-258
- 73 Vukelja S J, Baker W J, Jeffreys P. Nonbacterial thrombotic endocarditis clinically mimicking veno-occlusive disease of the liver complicating autologous bone marrow transplantation. Am J Clin Oncol . 1992; 15 500-502
- 74 McDonald G B, Sharma P, Matthews D E. The clinical course of 53 patients with venocclusive disease of the liver after marrow transplantation. Transplantation . 1985; 36 603-608
- 75 Bearman S I. The syndrome of hepatic venocclusive disease after marrow transplantation. Blood . 1995; 85 3005-3020
- 76 Wingard J R, Mellits E D, Jones R J. Association of hepatic veno-occlusive disease with interstitial pneumonitis in bone marrow transplant recipients. Bone Marrow Transplant . 1989; 4 685-689
- 77 Zager R A, O'Quigley J, Zager B K. Acute renal failure following bone marrow transplantation: A retrospective study of 272 patients. Am J Kidney Dis . 1989; 13 210-216
- 78 Bearman S I, Anderson G L, Mori M. Venocclusive disease of the liver: development of a model for predicting fatal outcome after marrow transplantation. J Clin Oncol . 1993; 11 1729-1736
- 79 Bearman S I, Lee J L, Baron A E. Treatment of hepatic venocclusive disease with recombinant human tissue plasminogen activator and heparin in 42 marrow transplant patients. Blood . 1997; 89 1501-1506
- 80 Vonnahme F-J. Die Leber des Menschen. Rasterelektronenmikroskopischer atlas. [The Human Liver. A Scanning Electron Microscopic Atlas, 1st ed.] Freiburg, Germany: Karger 1993
- 81 Shulman H M, Gown A M, Nugent D J. Hepatic veno-occlusive disease after bone marrow transplantation. Immunohistochemical identification of the material within occluded central venules. Am J Pathol . 1987; 127 549-558
- 82 Watanabe K, Iwaki H, Satoh M. Veno-occlusive disease of the liver following bone marrow transplantation: a clinical-pathological study of autopsy cases. Artif Organs . 1996; 20 1145-1150
- 83 Sato Y, Asada Y, Hara S. Hepatic stellate cells (Ito cells) in veno-occlusive disease of the liver after allogeneic bone marrow transplantation. Histopathology . 1999; 34 66-70
- 84 DeLeve L D, Wang X D, Huybrechts M M. Cellular target of cyclophosphamide toxicity in the murine liver-role of glutathione and site of metabolic activation. Hepatology . 1996; 24 830-837
- 85 DeLeve L D. Dacarbazine toxicity in murine liver cells: a model of hepatic endothelial injury and glutathione defense. J Pharmacol Exp Ther . 1994; 268 1261-1270
- 86 DeLeve L D, Ito Y, Machen N W. Embolization by sinusoidal lining cells causes the congestion of hepatic venocclusive disease. Gastroenterology (Abst) . 2000; 118 A1003
- 87 DeLeve L D, Wang X. Identification of two distinct mechanisms of busulfan toxicity in isolated murine hepatocytes. Hepatology . 1996; 24 464(Abst)
- 88 Wang X, Kanel G C, DeLeve L D. Support of sinusoidal endothelial cell glutathione prevents hepatic veno-occlusive disease in the rat. Hepatology . 2000; 31 428-434
- 89 Teicher B A, Crawford J M, Holden S A. Glutathione monoethyl ester can selectively protect liver from high dose BCNU or cyclophosphamide. Cancer . 1988; 62 1275-1281
- 90 DeLeve L D, Wang X, Tsai J. Prevention of hepatic venocclusive disease in the rat by inhibition of matrix metalloproteinases. Gastroenterology . 2001; 120 A54(Abst)
- 91 Cai T, Fassina G, Morini M. N-acetylcysteine inhibits endothelial cell invasion and angiogenesis. Lab Invest . 1999; 79 1151-1159
- 92 Tyagi S C, Ratajska A, Weber K T. Myocardial matrix metalloproteinase(s): localization and activation. Mol Cell Biochem . 1993; 126 49-59
- 93 Tyagi S C, Kumar S, Borders S. Reduction-oxidation (redox) state regulation of extracellular matrix metalloproteinases and tissue inhibitors in cardiac normal and transformed fibroblast cells. J Cell Biochem . 1996; 61 139-151
- 94 Upadhya G A, Strasberg S M. Glutathione, lactobionate, and histidine: cryptic inhibitors of matrix metalloproteinases contained in University of Wisconsin and histidine/tryptophan/ketoglutarate liver preservation solutions. Hepatology . 2000; 31 1115-1122
- 95 DeLeve L D, Wang X. Decrease in hepatic nitric oxide production contributes to hepatic veno-occlusive disease (HVOD). Hepatology . 1999; 30 218(Abst)
- 96 Eberhardt W, Beeg T, Beck K F. Nitric oxide modulates expression of matrix metalloproteinase-9 in rat mesangial cells. Kidney Int . 2000; 57 59-69
- 97 Korte W. Veno-occlusive disease of the liver after bone marrow transplantation: is hypercoagulability really part of the problem?. Blood Coagulation Fibrinolysis . 1997; 8 367-381
- 98 Brooks S E, Miller C G, McKenzie K. Acute veno-occlusive disease of the liver. Fine structure in Jamaican children. Arch Pathol Lab Med . 1970; 89 507-520
- 99 Radich J P, Sanders J E, Buckner C D. Second allogeneic marrow transplantation for patients with recurrent leukemia after initial transplant with TBI-containing regimens. J Clin Oncol . 1993; 11 304-313
- 100 Rollins B J. Hepatic veno-occlusive disease. Am J Med . 1986; 81 297-306
- 101 Slattery J T, Kalhorn T F, McDonald G B. Conditioning regimen-dependent disposition of cyclophosphamide and hydroxycyclophosphamide in human marrow transplantation patients. J Clin Oncol . 1996; 14 1484-1494
- 102 Honjo I, Suou T, Hirayama C. Hepatotoxicity of cyclophosphamide in man: Pharmacokinetic analysis. Res Commun Chem Pathol Pharmacol . 1988; 61 149-165
- 103 Gurtoo H L, Dahms R, Hipkens J. Studies of the binding of [3H-chloroethyl]-cyclophosphamide and 14[C-4]-cyclosphosphamide to hepatic microsomes and native calf thymus DNA. Life Sci . 1977; 22 45-52
- 104 Grochow L B, Piantadosi S, Santos G. Busulfan dose adjustment decreases the risk of hepatic veno-occlusive disease in patients undergoing bone marrow transplantation. Proc Am Assoc Cancer Res . 1992; 33 200(Abst)
- 105 Yeager A M, Wagner J E, Graham M L. Optimization of busulfan dosage in children undergoing bone marrow transplantation: a pharmacokinetic study of dose escalation. Blood . 1992; 80 2425-2428
- 106 Demirer T, Buckner C D, Appelbaum F R. Busulfan, cyclophosphamide and fractionated total body irradiation for allogeneic marrow transplantation for advanced acute and chronic myelogenous leukemia: phase I dose escalation of busulfan based on targeted plasma levels. Bone Marrow Transplant . 1996; 17 341-346
- 107 Dix S P, Wingard J R, Mullins R E. Association of busulfan area under the curve with veno-occlusive disease following BMT. Bone Marrow Transplant . 1996; 17 225-230
- 108 Vassal G, Deroussent A, Challine D. Is 600 mg/m2 the appropriate dosage of busulfan in children undergoing bone marrow transplantation?. Blood . 1992; 79 2475-2479
- 109 Schuler U, Schroer S, Kuhnle A. Busulfan pharmacokinetics in bone marrow transplant patients: is drug monitoring warranted?. Bone Marrow Transplant . 1994; 14 759-765
- 110 Slattery J T, Clift R A, Buckner C D. Marrow transplantation for chronic myeloid leukemia-the influence of plasma busulfan levels on the outcome of transplantation. Blood . 1997; 89 3055-3060
- 111 Shaw P J, Scharping C E, Brian R J. Busulfan pharmacokinetics using a single daily high-dose regimen in children with acute leukemia. Blood . 1994; 84 2357-2362
- 112 DeLeve L D, Wang X. Role of oxidative stress and glutathione in busulfan toxicity in cultured murine hepatocytes. Pharmacology . 2000; 60 143-154
- 113 Meresse V, Hartmann O, Vassal G. Risk factors of hepatic venocclusive disease after high-dose busulfan-containing regimens followed by autologous bone marrow transplantation: a study in 136 children. Bone Marrow Transplant . 1992; 10 135-141
- 114 Hassan M. The role of busulfan in bone marrow transplantation [Review]. Med Oncol . 1999; 16 166-176
- 115 Andersson B S, Gajewski J, Donato M. Allogeneic stem cell transplantation (BMT) for AML and MDS following i.v busulfan and cyclophosphamide (i.v. BuCy). Bone Marrow Transplant . 2000; 25(Suppl 2) 35-38
- 116 Fajardo L F, Colby T V. Pathogenesis of veno-occlusive liver disease after radiation. Arch Pathol Lab Med . 1980; 104 584-588
- 117 Girinsky T, Benhamou E, Bourhis J H. Prospective randomized comparison of single-dose versus hyperfractionated total-body irradiation in patients with hematologic malignancies. J Clin Oncol . 2000; 18 981-986
- 118 Ozsahin M, Pene F, Touboul E. Total-body irradiation before bone marrow transplantation. Results of two randomized instantaneous dose rates in 157 patients. Cancer . 1992; 69 2853-2865
- 119 Belkacemi Y, Ozsahin M, Rio B. Is veno-occlusive disease incidence influenced by the total-body irradiation technique?. Strahlenther Onkol . 1995; 171 694-697
- 120 Gordon B, Haire H, Kessinger A. High frequency of antithrombin 3 and protein C deficiency following autologous bone marrow transplantation for lymphoma. Bone Marrow Transplant . 1991; 8 497-502
- 121 Lee J H, Lee K H, Kim S. Relevance of proteins C and S, antithrombin III, von Willebrand factor, and Factor VIII for development of hepatic veno-occlusive disease in patients undergoing allogeneic bone marrow transplantation: a prospective study. Bone Marrow Transplantation . 1998; 22 883-888
- 122 Lee J H, Lee K H, Choi S J. Veno-occlusive disease of the liver after allogeneic bone marrow transplantation for severe aplastic anemia. Bone Marrow Transplant . 2000; 26 657-662
- 123 Salat C, Holler E, Kolb H J. The relevance of plasminogen activator inhibitor 1 (PAI-1) as a marker for the diagnosis of hepatic veno-occlusive disease in patients after bone marrow transplantation [Review]. Leuk Lymphoma . 1999; 33 25-32
- 124 Villalon L, Avello A G, Cesar J. Is veno-occlusive disease a specific syndrome or the exacerbation of physiopathologic hemostatic changes in hematopoietic stem cell transplantation (HSCT)?. Thromb Res . 2000; 99 439-446
- 125 Budinger M D, Bouvier M, Shah A. Results of a phase I trial of antithrombin III as prophylaxis in bone marrow transplant patients at risk for venocclusive disease [Abstract]. Blood . 1996; 88 172
- 126 Baglin T P, Harper P, Marcus R E. Venocclusive disease of the liver complicating ABMT successfully treated with recombinant tissue plasminogen activator. Bone Marrow Transplant . 1990; 5 439-441
- 127 Rosti G, Bandini G, Belardinelli A. Alteplase for hepatic veno-occlusive disease after bone-marrow transplantation. Lancet (Lett). 1992; 339 1481-1482
- 128 Laporte J P, Lesage S, Tilleul P. Alteplase for hepatic veno-occlusive disease complicating bone-marrow transplantation. Lancet . 1992; 339 1057(Lett)
- 129 Leahey A M, Bunin N J. Recombinant human tissue plasminogen activator for the treatment of severe hepatic veno-occlusive disease in pediatric bone marrow transplant patients. Bone Marrow Transplant . 1996; 17 1101-1104
- 130 Kulkarni S, Rodriguez M, Lafuente A. Recombinant tissue plasminogen activator (rtPA) for the treatment of hepatic veno-occlusive disease (VOD). Bone Marrow Transplantation . 1999; 23 803-807
- 131 Yoshimi A, Kato K, Maeda N. [Treatment of hepatic veno-occlusive disease after bone marrow transplantation with recombinant human tissue plasminogen activator (rh-tPA)] [in Japanese]. Rinsho Ketsueki . 2000; 41 103-108
- 132 Duggan C, Schmidt M, Lawler M. The prothrombin gene variant G20210A but not Factor V Leiden may be associated with veno-occlusive disease following BMT. Bone Marrow Transplant . 1999; 24 693-699
- 133 Chauncey T R, Thompson A R, Stewart L. No association between activated protein C (APC) resistance and hepatic veno-occlusive disease (VOD) following stem cell transplantation. Blood (Abst). 1997; 90 356-357
- 134 Schuppan D, Farrand A, Oesterling C. Circulating markers of hepatic fibrosis predict evolution of venocclusive disease after marrow transplantation. Hepatology . 1997; 26 452(Abst)
- 135 Friedman S L. Molecular regulation of hepatic fibrosis, an integrated cellular response to tissue injury. J Biol Chem . 2000; 275 2247-2250
- 136 Tanaka J, Imamura M, Kasai M. Rapid analysis of tumor necrosis factor-alpha mRNA expression during venocclusive disease of the liver after allogeneic bone marrow transplantation. Transplantation . 1993; 55 430-432
- 137 Ringden O, Remberger M, Lehmann S. N-acetylcysteine for hepatic veno-occlusive disease after allogeneic stem cell transplantation. Bone Marrow Transplant . 2000; 25 993-996
- 138 Farrand A L, Nash R A, Zager R A. Pathogenesis of venocclusive disease of the liver: role of cytokines, NO, endothelin-1, and fibrosis. Gastroenterology . 1996; 110 1189(Abst)
- 139 Rajvanshi R, Farrand A, Bouvier M E. Plasma levels of endothelin-1 and nitrate in patients with venocclusive disease of the liver after hematopoietic cell transplantation. Hepatology . 2000; 32 405(Abst)
- 140 Fink J, Cooper M, Burkhart K. Marked enzymuria following bone marrow transplantation: a correlate of veno-occlusive disease-induced ``hepatorenal syndrome''. J Am Soc Nephrol . 1995; 6 1655-1660
- 141 Lucarelli G, Clift R A, Galimberti M. Marrow transplantation for patients with thalassemia: results in class 3 patients. Blood . 1996; 87 2082-2088
- 142 Lucarelli G, Galimberti M, Polchi P. Bone marrow transplantation in patients with thalassemia. N Engl J Med . 1990; 322 417-421
- 143 Hassan M, Ehrsson H, Ljungman P. Aspects concerning busulfan pharmacokinetics and bioavailability. Leuk Lymphoma . 1996; 22 395-407
- 144 Demirer T, Buckner C D, Appelbaum F R. Busulfan, cyclophosphamide and fractionated total body irradiation for autologous or syngeneic marrow transplantation for acute and chronic myelogenous leukemia: phase I dose escalation of busulfan based on targeted plasma levels. Bone Marrow Transplant . 1996; 17 491-495
- 145 Locasciulli A, Bacigalupo A, Alberti A. Predictability before transplant of hepatic complications following allogeneic bone marrow transplantation. Transplantation . 1989; 48 68-72
- 146 Hagglund H, Remberger M, Klaesson S. Norethisterone treatment, a major risk factor for veno-occlusive disease in the liver after allogeneic bone marrow transplantation. Blood . 1998; 92 4568-4572
- 147 Ayash L J, Hunt M, Antman K. Hepatic venocclusive disease in autologous bone marrow transplantation of solid tumors and lymphomas. J Clin Oncol . 1990; 8 1699-1706
- 148 Anderson J E, Appelbaum F R, Schoch G. Relapse after allogeneic bone marrow transplantation for refractory anemia is increased by shielding lungs and liver during total body irradiation. Biol Blood Marrow Transplant . 2001; 7 163-170
- 149 Bearman S I, Hinds M S, Wolford J L. A pilot study of continuous infusion heparin for the prevention of hepatic venocclusive disease after bone marrow transplantation. Bone Marrow Transplant . 1990; 5 407-411
- 150 Attal M, Huguet F, Rubie H. Prevention of hepatic veno-occlusive disease after bone marrow transplantation by continuous infusion of low-dose heparin: a prospective, randomized trial. Blood . 1992; 79 1-7
- 151 Rosenthal J, Sender L, Secola R. Phase II trial of heparin prophylaxis for veno-occlusive disease of the liver in children undergoing bone marrow transplantation. Bone Marrow Transplant . 1996; 18 185-191
- 152 Marsa-Vila L, Gorin N C, Laporte J P. Prophylactic heparin does not prevent liver venocclusive disease following autologous bone marrow transplantation. Eur J Haematol . 1991; 47 346-354
- 153 Cahn J Y, Flesch M, Brion A. Prevention of veno-occlusive disease of the liver after bone marrow transplantation: heparin or no heparin?. Blood . 1992; 80 2149-2150
- 154 Or R, Nagler A, Shpilberg O. Low molecular weight heparin for the prevention of veno-occlusive disease of the liver in bone marrow transplantation patients. Transplantation . 1996; 61 1067-1071
- 155 Essell J H, Schroeder M T, Harman G S. Ursodiol prophylaxis against hepatic complications of allogeneic bone marrow transplantation. A randomized, double-blind, placebo-controlled trial. Ann Intern Med . 1998; 128 975-981
- 156 Ohashi K, Tanabe J, Watanabe R. The Japanese multicenter open randomized trial of ursodeoxycholic acid prophylaxis for hepatic veno-occlusive disease after stem cell transplantation. Am J Hematol . 2000; 64 32-38
- 157 Ruutu T, Eriksson B, Remes K. Ursodiol for the prevention of hepatic complications in allogeneic stem cell transplantation. Bone Marrow Transplant . 1999; 23 756(Abst)
- 158 Gluckman E, Jolivet I, Scrobohaci M L. Use of prostaglandin E1 for prevention of liver veno-occlusive disease in leukaemic patients treated by allogeneic bone marrow transplantation. Br J Haematol . 1990; 74 277-281
- 159 Bearman S I, Shen D, Hinds M S. A phase I/II study of prostaglandin E1 for the prevention of hepatic venocclusive disease after bone marrow transplantation. Br J Haematol . 1993; 84 724-730
- 160 Clift R A, Bianco J A, Appelbaum F R. A randomized controlled trial of pentoxifylline for the prevention of regimen-related toxicities in patients undergoing allogeneic marrow transplantation. Blood . 1993; 82 2025-2030
- 161 Attal M, Huguet F, Rubie H. Prevention of regimen-related toxicities after bone marrow transplantation by pentoxifylline: a prospective randomized trial. Blood . 1993; 82 732-736
- 162 Rubenfeld G D, Crawford S W. Withdrawing life support from mechanically ventilated recipients of bone marrow transplants: a case for evidence-based guidelines. Ann Intern Med . 1996; 125 625-633
- 163 Bearman S I, Shuhart M C, Hinds M S. Recombinant human tissue plasminogen activator for the treatment of established severe hepatic venocclusive disease of the liver after bone marrow transplantation. Blood . 1992; 80 2458-2462
- 164 Ringden O, Wennberg L, Ericzon B G. Alteplase for hepatic veno-occlusive disease after bone marrow transplantation [Letter]. Lancet . 1992; 340 546-547
- 165 Hagglund H, Ringden O, Ericzon B G. Treatment of hepatic venoocclusive disease with recombinant human tissue plasminogen activator or orthotopic liver transplantation after allogeneic bone marrow transplantation. Transplantation . 1996; 62 1076-1080
- 166 Palmer K J, Goa K L. Defibrotide. A review of its pharmacodynamic and pharmacokinetic properties, and therapeutic use in vascular disorders. Drugs . 1993; 45 259-294
- 167 Richardson P G, Elias A D, Krishnan A. Treatment of severe veno-occlusive disease with defibrotide: compassionate use results in efficacy without significant toxicity in a high risk population. Blood . 1998; 92 737-744
- 168 Chopra R, Eaton J D, Grassi A. Defibrotide for the treatment of hepatic veno-occlusive disease: results of the European compassionate-use study. Br J Haematol . 2000; 111 1122-1129
- 169 Haire W D, Ruby E I, Stephens L C. A prospective randomized double-blind trial of antithrombin III concentrate in the treatment of multiple-organ dysfunction syndrome during hematopoietic stem cell transplantation. Biol Blood Marrow Transplant . 1998; 4 142-150
- 170 Mertens R, Brost H, Granzen B. Antithrombin treatment of severe hepatic veno-occlusive disease in children with cancer. Eur J Pediatr . 1999; 158(Suppl 3) 154-158
- 171 Ibrahim A, Pico J L, Maraninchi D. Hepatic venocclusive disease following bone marrow transplantation treated by prostaglandin E1 . Bone Marrow Transplant . 1991; 7(Suppl 2) 53
- 172 Khoury H, Adkins D, Brown R. Does early treatment with high-dose methylprednisolone alter the course of hepatic regimen-related toxicity?. Bone Marrow Transplant . 2000; 25 737-743
- 173 Kajiume T, Yoshimi S, Nagita A. Application of nitric oxide for a case of veno-occlusive disease after peripheral blood stem cell transplantation. Paediatr Hematol Oncol . 2000; 17 601-604
- 174 Nattakom T V, Charlton A, Wilmore D W. Use of vitamin E and glutamine in the successful treatment of severe veno-occlusive disease following bone marrow transplantation. Nutr Clin Pract . 1995; 10 16-18
- 175 Smith F O, Johnson M S, Scherer L R. Transjugular intrahepatic portosystemic shunting (TIPS) for treatment of severe hepatic veno-occlusive disease. Bone Marrow Transplant . 1996; 18 643-646
- 176 Fried M W, Connaghan D G, Sharma S. Transjugular intrahepatic portosystemic shunt for the management of severe venoocclusive disease following bone marrow transplantation. Hepatology . 1996; 24 588-591
- 177 Azoulay D, Castaing D, Lemoine A. Transjugular intrahepatic portosystemic shunts (TIPS) for severe veno-occlusive disease of the liver following bone marrow transplantation. Bone Marrow Transplant . 2000; 25 987-992
- 178 Zenz T, Rossle M, Bertz H. Severe veno-occlusive disease after allogeneic bone marrow or peripheral stem cell transplantation-role of transjugular intrahepatic portosystemic shunt (TIPS). Liver . 2001; 21 31-36
- 179 Rajvanshi P, McDonald G B. Expanding the use of transjugular intrahepatic portosystemic shunts for veno-occlusive disease. Liver Transplant . 2001; 7 154-159
- 180 Meacher R, Venkatesh B, Lipman J. Acute respiratory distress syndrome precipitated by transjugular intrahepatic porto-systemic shunting for severe hepatic veno-occlusive disease. Is it due to pulmonary leucostasis?. Intensive Care Med . 1999; 25 1332-1333
- 181 Murray J A, LaBrecque D R, Gingrich R D. Successful treatment of hepatic venocclusive disease in a bone marrow transplant patient with side-to-side portacaval shunt. Gastroenterology . 1987; 92 1073-1077
- 182 Jacobson B K, Kalayoglu M. Effective early treatment of hepatic venocclusive disease with a central splenorenal shunt in an infant. J Pediatr Surg . 1992; 27 531-533
- 183 Nimer S D, Milewicz A L, Champlin R E. Successful treatment of hepatic venocclusive disease in a bone marrow transplant patient with orthotopic liver transplantation. Transplantation . 1990; 49 819-821
- 184 Rapoport A P, Doyle H R, Starzl T. Orthotopic liver transplantation for life-threatening veno-occlusive disease of the liver after allogeneic bone marrow transplant. Bone Marrow Transplant . 1991; 8 421-424
- 185 Salat C, Holler E, Wolf C. Laboratory markers of veno-occlusive disease in the course of bone marrow and subsequent liver transplantation. Bone Marrow Transplant . 1997; 19 487-490
- 186 Schlitt H J, Tischler H J, Ringe B. Allogeneic liver transplantation for hepatic veno-occlusive disease after bone marrow transplantation-clinical and immunological considerations. Bone Marrow Transplant . 1995; 16 473-478
- 187 Dowlati A, Honore P, Damas P. Hepatic rejection after orthotopic liver transplantation for hepatic veno-occlusive disease or graft-versus-host disease following bone marrow transplantation. Transplantation . 1995; 60 106-109
- 188 Bunin N, Leahey A, Dunn S. Related donor liver transplant for veno-occlusive disease following T-depleted unrelated donor bone marrow transplantation. Transplantation . 1996; 61 664-666
- 189 Rosen H R, Martin P, Schiller G J. Orthotopic liver transplantation for bone-marrow transplant-associated veno-occlusive disease and graft-versus-host disease of the liver. Liver Transplant Surg . 1996; 2 225-232
REFERENCES
- 1 Willmot F C, Robertson G W. Senecio disease, or cirrhosis of the liver due to senecio poisoning. Lancet . 1920; 2 848-849
- 2 Selzer G, Parker R GF. Senecio poisoning exhibiting as Chiari's syndrome: a report on twelve cases. Am J Pathol . 1950; 27 885-887
- 3 Bras G, Jeliffe D B, Stuart K L. Veno-occlusive disease of the liver with non-portal type of cirrhosis occurring in Jamaica. Arch Pathol . 1954; 57 285-300
- 4 DeLeve L D, McCuskey R S, Wang X. Characterization of a reproducible rat model of hepatic veno-occlusive disease. Hepatology . 1999; 29 1779-1791
- 5 Shulman H M, Fisher L B, Schoch H G. Venocclusive disease of the liver after marrow transplantation: histologic correlates of clinical signs and symptoms. Hepatology . 1994; 19 1171-1180
- 6 Mohabbat O, Younos M S, Merzad A A. An outbreak of hepatic veno-occlusive disease in north-western Afghanistan. Lancet . 1976; 2 269-271
- 7 Tandon H D, Tandon B N, Mattocks A R. An epidemic of veno-occlusive disease of the liver in Afghanistan. Pathologic features. Am J Gastroenterol . 1978; 70 607-613
- 8 Eisenhauer T, Hartmann H, Rumpf K W. Favourable outcome of hepatic veno-occlusive disease in a renal transplant patient receiving azathioprine, treated by portacaval shunt. Report of a case and review of the literature. Digestion . 1984; 30 185-190
- 9 Katzka D A, Saul S H, Jorkasky D. Azathioprine and hepatic venocclusive disease in renal transplant patients. Gastroenterology . 1986; 90 446-454
- 10 Read A E, Weisner R H, LaBrecque D R. Hepatic venocclusive disease associated with renal transplantation and azathioprine therapy. Ann Intern Med . 1986; 104 651-655
- 11 Haboubi N Y, Ali H H, Whitwell H L. Role of endothelial cell injury in the spectrum of azathioprine-induced liver disease after renal transplant: light microscopy and ultrastructural observations. Am J Gastroenterol . 1988; 83 256-261
- 12 Liano F, Moreno A, Matesanz R. Veno-occlusive hepatic disease of the liver in renal transplantation: is azathioprine the cause?. Nephron . 1989; 51 509-516
- 13 Sterneck M, Wiesner R, Ascher N. Azathioprine hepatotoxicity after liver transplantation [Review]. Hepatology . 1991; 14 806-810
- 14 de Araujo S M, Gerard F, Chossegros P. Vascular hepatotoxicity related to heroin addiction. Virchows Arch [A] . 1990; 417 497-503
- 15 Trigueiro de Araujo S M, Gerard F, Chossegros P. Lack of hepatocyte involvement in the genesis of the sinusoidal dilatation related to heroin addiction: a morphometric study. Virchows Arch [A] . 1992; 420 149-153
- 16 Zafrani E S, Cazier A, Baudelot A M. Ultrastructural lesions of the liver in human peliosis. A report of 12 cases. Am J Pathol . 1984; 114 349-359
- 17 DeLeve L D, Wang X, Kuhlenkamp J F. Toxicity of azathioprine and monocrotaline in murine sinusoidal endothelial cells and hepatocytes: the role of glutathione and relevance to hepatic veno-occlusive disease. Hepatology . 1996; 23 589-599
- 18 DeLeve L D. Glutathione defense in non-parenchymal cells [Review]. Semin Liver Dis . 1998; 18 403-413
- 19 Giles F J, Kantarjian H M, Kornblau S M. Mylotarg (gemtuzumab ozogamicin) therapy is associated with hepatic venoocclusive disease in patients who have not received stem cell transplantation. Cancer . 2001; 92 406-413
- 20 Rajvanshi P, Shulman H M, Sievers E L. Hepatic sinusoidal obstruction following Gemtuzumab ozogamicin (Mylotarg). Blood (in press). 2002;
- 21 King P D, Perry M C. Hepatotoxicity of chemotherapy [Review]. Oncologist . 2001; 6 162-176
- 22 Tornesello A, Piciacchia D, Mastrangelo S. Veno-occlusive disease of the liver in right-sided Wilms' tumours [Review]. Eur J Cancer . 1998; 34 1220-1223
- 23 Czauderna P, Katski K, Kowalczyk J. Venoocclusive liver disease (VOD) as a complication of Wilms' tumour management in the series of consecutive 206 patients. Eur J Pediatr Surg . 2000; 10 300-303
- 24 Lawrence T S, Robertson J M, Anscher M S. Hepatic toxicity resulting from cancer treatment [Review]. Int J Radiat Oncol Biol Phys . 1995; 31 1237-1248
- 25 Vowels M, Lam-Po-Tang R, Zagars G. Total body irradiation and Budd-Chiari syndrome. Pathology . 1979; 11 306
- 26 Berk P D, Popper H, Krueger G RF. Veno-occlusive disease of the liver after allogeneic bone marrow transplantation. Ann Intern Med . 1979; 90 158-164
- 27 Jacobs P, Miller J L, Uys C J. Fatal veno-occlusive disease of the liver after chemotherapy, whole-body irradiation and bone marrow transplantation for refractory acute leukaemia. S Afr Med J . 1979; 55 5-10
- 28 McDonald G B, Hinds M S, Fisher L B. Venocclusive disease of the liver and multiorgan failure after bone marrow transplantation: a cohort study of 355 patients. Ann Intern Med . 1993; 118 255-267
- 29 McDonald G B, Sharma P, Matthews D E. Venocclusive disease of the liver after bone marrow transplantation: diagnosis, incidence, and predisposing factors. Hepatology . 1984; 4 116-122
- 30 Ganem G, Saint-Marc Girardin F M, Kuentz M. Venocclusive disease of the liver after allogeneic bone marrow transplantation in man. Int J Radiat Oncol Biol Phys . 1988; 14 879-884
- 31 Jones R J, Lee K S, Beschorner W E. Veno-occlusive disease of the liver following bone marrow transplantation. Transplantation . 1987; 44 778-783
- 32 Carreras E, Bertz H, Arcese W. Incidence and outcome of hepatic veno-occlusive disease after blood or marrow transplantation: a prospective cohort study of the European group for blood and marrow transplantation. Blood . 1998; 92 3599-3604
- 33 Hasegawa S, Horibe K, Kawabe T. Veno-occlusive disease of the liver after allogeneic bone marrow transplantation in children with hematologic malignancies: incidence, onset time and risk factors. Bone Marrow Transplant . 1998; 22 1191-1197
- 34 McSweeney P A, Niederwieser D, Shizuru J A. Hematopoietic cell transplantation in older patients with hematologic malignancies: replacing high-dose cytotoxic therapy with graft-versus-tumor effects. Blood . 2001; 97 3390-3400
- 35 McDonald G B, Ren S, Bouvier M E. Venocclusive disease of the liver and cyclophosphamide pharmacokinetics: a prospective study in marrow transplant patients [Abstract]. Hepatology . 1999; 30 314
- 36 Strasser S I, Myerson D, Spurgeon C L. Hepatitis C virus infection after bone marrow transplantation: a cohort study with 10 year follow-up. Hepatology . 1999; 29 1893-1899
- 37 Hagglund H, Remberger M, Klaesson S. Norethisterone treatment, a major risk factor for veno-occlusive disease in the liver after allogeneic bone marrow transplantation. Blood . 1998; 92 4568-4572
- 38 Lee J L, Gooley T, Bensinger W. Venocclusive disease of the liver after busulfan, melphalan, and thioTEPA conditioning therapy: incidence, risk factors, and outcome. Biol Blood Marrow Transplantation . 1999; 5 306-315
- 39 Toh H C, McAfee S L, Sackstein R. Late onset veno-occlusive disease following high-dose chemotherapy and stem cell transplantation. Bone Marrow Transplant . 1999; 24 891-895
- 40 Strasser S I, McDonald G B. Hepatobiliary complications of hematopoietic stem cell transplantation. In: Schiff ER, Sorrell MF, Maddrey WC, eds. Schiff's Diseases of the Liver, 8th ed Philadelphia: Lippincott-Raven, 1999: 1617-1641
- 41 Shulman H M, McDonald G B, Matthews D. An analysis of hepatic venocclusive disease and centrilobular hepatic degeneration following bone marrow transplantation. Gastroenterology . 1980; 79 1178-1191
- 42 Strasser S I, McDonald S J, Schoch H G. Severe hepatocellular injury after hematopoietic cell transplant: incidence and etiology in 2136 consecutive patients [Abstract]. Hepatology . 2000; 32 299
- 43 Blann A, Collins P. Von Willebrand factor, angiotensin converting enzyme and endothelial cell damage in veno-occlusive disease and bone marrow transplantation. Thromb Res . 1992; 66 617-618
- 44 Holler E, Kolb H J, Moller A. Increased serum levels of tumor necrosis factor-a precede major complications of bone marrow transplantation. Blood . 1990; 75 1011-1016
- 45 Anscher M S, Peters W P, Reisenbichler H. Transforming growth factor βas a predictor of liver and lung fibrosis after autologous bone marrow transplantation for advanced breast cancer. N Engl J Med . 1993; 328 1592-1598
- 46 Oh H, Tahara T, Bouvier M. Plasma thrombopoietin levels in marrow transplant patients with veno-occlusive disease of the liver. Bone Marrow Transplant . 1998; 22 675-679
- 47 Eltumi M, Trivedi P, Hobbs J. Monitoring of veno-occlusive disease after bone marrow transplantation by serum aminopropeptide of type III procollagen. Lancet . 1993; 342 518-521
- 48 Rio B, Bauduer F, Arrago J P. N-terminal peptide of type III procollagen: a marker for the development of hepatic veno-occlusive disease after BMT and a basis for determining the timing of prophylactic heparin. Bone Marrow Transplant . 1993; 11 471-472
- 49 Park Y D, Yasui M, Yoshimoto T. Changes in hemostatic parameters in hepatic veno-occlusive disease following bone marrow transplantation. Bone Marrow Transplant . 1997; 19 915-920
- 50 Murase T, Jirtle R L, McDonald G B. Transforming growth factor-beta plasma concentrations in patients with leukemia and lymphoma receiving chemoradiotherapy and marrow transplantation [Letter]. Blood . 1994; 83 2383
- 51 Farrand A L, Bouvier M E, McDonald G B. Plasma levels of the propeptide of type III collagen predict the severity of venocclusive disease of the liver [Abstract]. Hepatology . 1996; 24 508
- 52 Heikinheimo M, Halila R, Fasth A. Serum procollagen type III is an early and sensitive marker for venocclusive disease of the liver in children undergoing bone marrow transplantation. Blood . 1994; 83 3036-3040
- 53 Salat C, Holler E, Kolb H J. Plasminogen activator inhibitor-1 confirms the diagnosis of hepatic veno-occlusive disease in patients with hyperbilirubinemia after bone marrow transplantation. Blood . 1997; 89 2184-2188
- 54 Remberger M, Ringden O. Increased levels of soluble interleukin-2 receptor in veno-occlusive disease of the liver after allogenic bone marrow transplantation. Transplantation . 1995; 60 1293-1299
- 55 Remberger M, Ringden O. Serum levels of cytokines after bone marrow transplantation: increased IL-8 levels during severe veno-occlusive disease of the liver. Eur J Haematol . 1997; 59 254-262
- 56 Scrobohaci M L, Drouet L, Monem-Mansi A. Liver venocclusive disease after bone marrow transplantation. Changes in coagulation parameters and endothelial markers. Thromb Res . 1991; 63 509-519
- 57 Faioni E M, Krachmalnicoff A, Bearman S I. Naturally occurring anticoagulants and bone marrow transplantation: plasma protein C predicts the development of venocclusive disease of the liver. Blood . 1993; 81 3458-3462
- 58 Rio B, Andreu G, Nicod A. Thrombocytopenia in venocclusive disease after bone marrow transplantation or chemotherapy. Blood . 1986; 67 1773-1776
- 59 Hommeyer S C, Teffey S A, Jacobson A F. Venocclusive disease of the liver: prospective study of US evaluation. Radiology . 1992; 184 683-686
- 60 Herbetko J, Grigg A P, Buckley A R. Veno-occlusive liver disease after bone marrow transplantation: findings at duplex sonography. AJR . 1992; 158 1001-1005
- 61 Lassau N, Leclere J, Auperin A. Hepatic veno-occlusive disease after myeloablative treatment and bone marrow transplantation: value of gray-scale and Doppler US in 100 patients. Radiology . 1997; 204 545-552
- 62 Costa F, Choy C G, Seiter K. Hepatic outflow obstruction and liver failure due to leukemic cell infiltration in chronic lymphocytic leukemia. Leuk Lymphoma . 1998; 30 403-410
- 63 Brown B P, Abu-Yousef M, Farner R. Doppler sonography: a noninvasive method for evaluation of hepatic venocclusive disease. AJR . 1990; 154 721-724
- 64 Deeg K H, Glockel U, Richter R. Diagnosis of veno-occlusive disease of the liver by color-coded Doppler sonography. Pediatr Radiol . 1993; 23 134-136
- 65 Nicolau C, Bru C, Carreras E. Sonographic diagnosis and hemodynamic correlation in veno-occlusive disease of the liver. J Ultrasound Med . 1993; 12 437-440
- 66 Grigg A, Gibson R, Bardy P. Acute portal vein thrombosis after autologous stem cell transplantation. Bone Marrow Transplant . 1996; 18 949-953
- 67 Kikuchi K, Rudolph R, Murakami C. Portal vein thrombosis after hematopoietic cell transplantation: frequency, treatment, and outcome. Bone Marrow Transplantation (in press). 2002;
- 68 McCarville M B, Hoffer F A, Howard S C. Hepatic veno-occlusive disease in children undergoing bone-marrow transplantation: usefulness of sonographic findings. Pediatr Radiol . 2001; 31 102-105
- 69 Carreras E, Granena A, Navasa M. Transjugular liver biopsy in bone marrow transplantation. Bone Marrow Transplant . 1993; 11 21-26
- 70 Shulman H M, Gooley T, Dudley M D. Utility of transvenous liver biopsies and wedged hepatic venous pressure measurements in sixty marrow transplant recipients. Transplantation . 1995; 59 1015-1022
- 71 Sharma P, McDonald G B, Banaji M. The risk of bleeding after percutaneous liver biopsy: relation to platelet count. J Clin Gastroenterol . 1982; 4 451-453
- 72 Zimmerman H J, Fang M, Utili R. Jaundice due to bacterial infection. Gastroenterology . 1979; 28 249-258
- 73 Vukelja S J, Baker W J, Jeffreys P. Nonbacterial thrombotic endocarditis clinically mimicking veno-occlusive disease of the liver complicating autologous bone marrow transplantation. Am J Clin Oncol . 1992; 15 500-502
- 74 McDonald G B, Sharma P, Matthews D E. The clinical course of 53 patients with venocclusive disease of the liver after marrow transplantation. Transplantation . 1985; 36 603-608
- 75 Bearman S I. The syndrome of hepatic venocclusive disease after marrow transplantation. Blood . 1995; 85 3005-3020
- 76 Wingard J R, Mellits E D, Jones R J. Association of hepatic veno-occlusive disease with interstitial pneumonitis in bone marrow transplant recipients. Bone Marrow Transplant . 1989; 4 685-689
- 77 Zager R A, O'Quigley J, Zager B K. Acute renal failure following bone marrow transplantation: A retrospective study of 272 patients. Am J Kidney Dis . 1989; 13 210-216
- 78 Bearman S I, Anderson G L, Mori M. Venocclusive disease of the liver: development of a model for predicting fatal outcome after marrow transplantation. J Clin Oncol . 1993; 11 1729-1736
- 79 Bearman S I, Lee J L, Baron A E. Treatment of hepatic venocclusive disease with recombinant human tissue plasminogen activator and heparin in 42 marrow transplant patients. Blood . 1997; 89 1501-1506
- 80 Vonnahme F-J. Die Leber des Menschen. Rasterelektronenmikroskopischer atlas. [The Human Liver. A Scanning Electron Microscopic Atlas, 1st ed.] Freiburg, Germany: Karger 1993
- 81 Shulman H M, Gown A M, Nugent D J. Hepatic veno-occlusive disease after bone marrow transplantation. Immunohistochemical identification of the material within occluded central venules. Am J Pathol . 1987; 127 549-558
- 82 Watanabe K, Iwaki H, Satoh M. Veno-occlusive disease of the liver following bone marrow transplantation: a clinical-pathological study of autopsy cases. Artif Organs . 1996; 20 1145-1150
- 83 Sato Y, Asada Y, Hara S. Hepatic stellate cells (Ito cells) in veno-occlusive disease of the liver after allogeneic bone marrow transplantation. Histopathology . 1999; 34 66-70
- 84 DeLeve L D, Wang X D, Huybrechts M M. Cellular target of cyclophosphamide toxicity in the murine liver-role of glutathione and site of metabolic activation. Hepatology . 1996; 24 830-837
- 85 DeLeve L D. Dacarbazine toxicity in murine liver cells: a model of hepatic endothelial injury and glutathione defense. J Pharmacol Exp Ther . 1994; 268 1261-1270
- 86 DeLeve L D, Ito Y, Machen N W. Embolization by sinusoidal lining cells causes the congestion of hepatic venocclusive disease. Gastroenterology (Abst) . 2000; 118 A1003
- 87 DeLeve L D, Wang X. Identification of two distinct mechanisms of busulfan toxicity in isolated murine hepatocytes. Hepatology . 1996; 24 464(Abst)
- 88 Wang X, Kanel G C, DeLeve L D. Support of sinusoidal endothelial cell glutathione prevents hepatic veno-occlusive disease in the rat. Hepatology . 2000; 31 428-434
- 89 Teicher B A, Crawford J M, Holden S A. Glutathione monoethyl ester can selectively protect liver from high dose BCNU or cyclophosphamide. Cancer . 1988; 62 1275-1281
- 90 DeLeve L D, Wang X, Tsai J. Prevention of hepatic venocclusive disease in the rat by inhibition of matrix metalloproteinases. Gastroenterology . 2001; 120 A54(Abst)
- 91 Cai T, Fassina G, Morini M. N-acetylcysteine inhibits endothelial cell invasion and angiogenesis. Lab Invest . 1999; 79 1151-1159
- 92 Tyagi S C, Ratajska A, Weber K T. Myocardial matrix metalloproteinase(s): localization and activation. Mol Cell Biochem . 1993; 126 49-59
- 93 Tyagi S C, Kumar S, Borders S. Reduction-oxidation (redox) state regulation of extracellular matrix metalloproteinases and tissue inhibitors in cardiac normal and transformed fibroblast cells. J Cell Biochem . 1996; 61 139-151
- 94 Upadhya G A, Strasberg S M. Glutathione, lactobionate, and histidine: cryptic inhibitors of matrix metalloproteinases contained in University of Wisconsin and histidine/tryptophan/ketoglutarate liver preservation solutions. Hepatology . 2000; 31 1115-1122
- 95 DeLeve L D, Wang X. Decrease in hepatic nitric oxide production contributes to hepatic veno-occlusive disease (HVOD). Hepatology . 1999; 30 218(Abst)
- 96 Eberhardt W, Beeg T, Beck K F. Nitric oxide modulates expression of matrix metalloproteinase-9 in rat mesangial cells. Kidney Int . 2000; 57 59-69
- 97 Korte W. Veno-occlusive disease of the liver after bone marrow transplantation: is hypercoagulability really part of the problem?. Blood Coagulation Fibrinolysis . 1997; 8 367-381
- 98 Brooks S E, Miller C G, McKenzie K. Acute veno-occlusive disease of the liver. Fine structure in Jamaican children. Arch Pathol Lab Med . 1970; 89 507-520
- 99 Radich J P, Sanders J E, Buckner C D. Second allogeneic marrow transplantation for patients with recurrent leukemia after initial transplant with TBI-containing regimens. J Clin Oncol . 1993; 11 304-313
- 100 Rollins B J. Hepatic veno-occlusive disease. Am J Med . 1986; 81 297-306
- 101 Slattery J T, Kalhorn T F, McDonald G B. Conditioning regimen-dependent disposition of cyclophosphamide and hydroxycyclophosphamide in human marrow transplantation patients. J Clin Oncol . 1996; 14 1484-1494
- 102 Honjo I, Suou T, Hirayama C. Hepatotoxicity of cyclophosphamide in man: Pharmacokinetic analysis. Res Commun Chem Pathol Pharmacol . 1988; 61 149-165
- 103 Gurtoo H L, Dahms R, Hipkens J. Studies of the binding of [3H-chloroethyl]-cyclophosphamide and 14[C-4]-cyclosphosphamide to hepatic microsomes and native calf thymus DNA. Life Sci . 1977; 22 45-52
- 104 Grochow L B, Piantadosi S, Santos G. Busulfan dose adjustment decreases the risk of hepatic veno-occlusive disease in patients undergoing bone marrow transplantation. Proc Am Assoc Cancer Res . 1992; 33 200(Abst)
- 105 Yeager A M, Wagner J E, Graham M L. Optimization of busulfan dosage in children undergoing bone marrow transplantation: a pharmacokinetic study of dose escalation. Blood . 1992; 80 2425-2428
- 106 Demirer T, Buckner C D, Appelbaum F R. Busulfan, cyclophosphamide and fractionated total body irradiation for allogeneic marrow transplantation for advanced acute and chronic myelogenous leukemia: phase I dose escalation of busulfan based on targeted plasma levels. Bone Marrow Transplant . 1996; 17 341-346
- 107 Dix S P, Wingard J R, Mullins R E. Association of busulfan area under the curve with veno-occlusive disease following BMT. Bone Marrow Transplant . 1996; 17 225-230
- 108 Vassal G, Deroussent A, Challine D. Is 600 mg/m2 the appropriate dosage of busulfan in children undergoing bone marrow transplantation?. Blood . 1992; 79 2475-2479
- 109 Schuler U, Schroer S, Kuhnle A. Busulfan pharmacokinetics in bone marrow transplant patients: is drug monitoring warranted?. Bone Marrow Transplant . 1994; 14 759-765
- 110 Slattery J T, Clift R A, Buckner C D. Marrow transplantation for chronic myeloid leukemia-the influence of plasma busulfan levels on the outcome of transplantation. Blood . 1997; 89 3055-3060
- 111 Shaw P J, Scharping C E, Brian R J. Busulfan pharmacokinetics using a single daily high-dose regimen in children with acute leukemia. Blood . 1994; 84 2357-2362
- 112 DeLeve L D, Wang X. Role of oxidative stress and glutathione in busulfan toxicity in cultured murine hepatocytes. Pharmacology . 2000; 60 143-154
- 113 Meresse V, Hartmann O, Vassal G. Risk factors of hepatic venocclusive disease after high-dose busulfan-containing regimens followed by autologous bone marrow transplantation: a study in 136 children. Bone Marrow Transplant . 1992; 10 135-141
- 114 Hassan M. The role of busulfan in bone marrow transplantation [Review]. Med Oncol . 1999; 16 166-176
- 115 Andersson B S, Gajewski J, Donato M. Allogeneic stem cell transplantation (BMT) for AML and MDS following i.v busulfan and cyclophosphamide (i.v. BuCy). Bone Marrow Transplant . 2000; 25(Suppl 2) 35-38
- 116 Fajardo L F, Colby T V. Pathogenesis of veno-occlusive liver disease after radiation. Arch Pathol Lab Med . 1980; 104 584-588
- 117 Girinsky T, Benhamou E, Bourhis J H. Prospective randomized comparison of single-dose versus hyperfractionated total-body irradiation in patients with hematologic malignancies. J Clin Oncol . 2000; 18 981-986
- 118 Ozsahin M, Pene F, Touboul E. Total-body irradiation before bone marrow transplantation. Results of two randomized instantaneous dose rates in 157 patients. Cancer . 1992; 69 2853-2865
- 119 Belkacemi Y, Ozsahin M, Rio B. Is veno-occlusive disease incidence influenced by the total-body irradiation technique?. Strahlenther Onkol . 1995; 171 694-697
- 120 Gordon B, Haire H, Kessinger A. High frequency of antithrombin 3 and protein C deficiency following autologous bone marrow transplantation for lymphoma. Bone Marrow Transplant . 1991; 8 497-502
- 121 Lee J H, Lee K H, Kim S. Relevance of proteins C and S, antithrombin III, von Willebrand factor, and Factor VIII for development of hepatic veno-occlusive disease in patients undergoing allogeneic bone marrow transplantation: a prospective study. Bone Marrow Transplantation . 1998; 22 883-888
- 122 Lee J H, Lee K H, Choi S J. Veno-occlusive disease of the liver after allogeneic bone marrow transplantation for severe aplastic anemia. Bone Marrow Transplant . 2000; 26 657-662
- 123 Salat C, Holler E, Kolb H J. The relevance of plasminogen activator inhibitor 1 (PAI-1) as a marker for the diagnosis of hepatic veno-occlusive disease in patients after bone marrow transplantation [Review]. Leuk Lymphoma . 1999; 33 25-32
- 124 Villalon L, Avello A G, Cesar J. Is veno-occlusive disease a specific syndrome or the exacerbation of physiopathologic hemostatic changes in hematopoietic stem cell transplantation (HSCT)?. Thromb Res . 2000; 99 439-446
- 125 Budinger M D, Bouvier M, Shah A. Results of a phase I trial of antithrombin III as prophylaxis in bone marrow transplant patients at risk for venocclusive disease [Abstract]. Blood . 1996; 88 172
- 126 Baglin T P, Harper P, Marcus R E. Venocclusive disease of the liver complicating ABMT successfully treated with recombinant tissue plasminogen activator. Bone Marrow Transplant . 1990; 5 439-441
- 127 Rosti G, Bandini G, Belardinelli A. Alteplase for hepatic veno-occlusive disease after bone-marrow transplantation. Lancet (Lett). 1992; 339 1481-1482
- 128 Laporte J P, Lesage S, Tilleul P. Alteplase for hepatic veno-occlusive disease complicating bone-marrow transplantation. Lancet . 1992; 339 1057(Lett)
- 129 Leahey A M, Bunin N J. Recombinant human tissue plasminogen activator for the treatment of severe hepatic veno-occlusive disease in pediatric bone marrow transplant patients. Bone Marrow Transplant . 1996; 17 1101-1104
- 130 Kulkarni S, Rodriguez M, Lafuente A. Recombinant tissue plasminogen activator (rtPA) for the treatment of hepatic veno-occlusive disease (VOD). Bone Marrow Transplantation . 1999; 23 803-807
- 131 Yoshimi A, Kato K, Maeda N. [Treatment of hepatic veno-occlusive disease after bone marrow transplantation with recombinant human tissue plasminogen activator (rh-tPA)] [in Japanese]. Rinsho Ketsueki . 2000; 41 103-108
- 132 Duggan C, Schmidt M, Lawler M. The prothrombin gene variant G20210A but not Factor V Leiden may be associated with veno-occlusive disease following BMT. Bone Marrow Transplant . 1999; 24 693-699
- 133 Chauncey T R, Thompson A R, Stewart L. No association between activated protein C (APC) resistance and hepatic veno-occlusive disease (VOD) following stem cell transplantation. Blood (Abst). 1997; 90 356-357
- 134 Schuppan D, Farrand A, Oesterling C. Circulating markers of hepatic fibrosis predict evolution of venocclusive disease after marrow transplantation. Hepatology . 1997; 26 452(Abst)
- 135 Friedman S L. Molecular regulation of hepatic fibrosis, an integrated cellular response to tissue injury. J Biol Chem . 2000; 275 2247-2250
- 136 Tanaka J, Imamura M, Kasai M. Rapid analysis of tumor necrosis factor-alpha mRNA expression during venocclusive disease of the liver after allogeneic bone marrow transplantation. Transplantation . 1993; 55 430-432
- 137 Ringden O, Remberger M, Lehmann S. N-acetylcysteine for hepatic veno-occlusive disease after allogeneic stem cell transplantation. Bone Marrow Transplant . 2000; 25 993-996
- 138 Farrand A L, Nash R A, Zager R A. Pathogenesis of venocclusive disease of the liver: role of cytokines, NO, endothelin-1, and fibrosis. Gastroenterology . 1996; 110 1189(Abst)
- 139 Rajvanshi R, Farrand A, Bouvier M E. Plasma levels of endothelin-1 and nitrate in patients with venocclusive disease of the liver after hematopoietic cell transplantation. Hepatology . 2000; 32 405(Abst)
- 140 Fink J, Cooper M, Burkhart K. Marked enzymuria following bone marrow transplantation: a correlate of veno-occlusive disease-induced ``hepatorenal syndrome''. J Am Soc Nephrol . 1995; 6 1655-1660
- 141 Lucarelli G, Clift R A, Galimberti M. Marrow transplantation for patients with thalassemia: results in class 3 patients. Blood . 1996; 87 2082-2088
- 142 Lucarelli G, Galimberti M, Polchi P. Bone marrow transplantation in patients with thalassemia. N Engl J Med . 1990; 322 417-421
- 143 Hassan M, Ehrsson H, Ljungman P. Aspects concerning busulfan pharmacokinetics and bioavailability. Leuk Lymphoma . 1996; 22 395-407
- 144 Demirer T, Buckner C D, Appelbaum F R. Busulfan, cyclophosphamide and fractionated total body irradiation for autologous or syngeneic marrow transplantation for acute and chronic myelogenous leukemia: phase I dose escalation of busulfan based on targeted plasma levels. Bone Marrow Transplant . 1996; 17 491-495
- 145 Locasciulli A, Bacigalupo A, Alberti A. Predictability before transplant of hepatic complications following allogeneic bone marrow transplantation. Transplantation . 1989; 48 68-72
- 146 Hagglund H, Remberger M, Klaesson S. Norethisterone treatment, a major risk factor for veno-occlusive disease in the liver after allogeneic bone marrow transplantation. Blood . 1998; 92 4568-4572
- 147 Ayash L J, Hunt M, Antman K. Hepatic venocclusive disease in autologous bone marrow transplantation of solid tumors and lymphomas. J Clin Oncol . 1990; 8 1699-1706
- 148 Anderson J E, Appelbaum F R, Schoch G. Relapse after allogeneic bone marrow transplantation for refractory anemia is increased by shielding lungs and liver during total body irradiation. Biol Blood Marrow Transplant . 2001; 7 163-170
- 149 Bearman S I, Hinds M S, Wolford J L. A pilot study of continuous infusion heparin for the prevention of hepatic venocclusive disease after bone marrow transplantation. Bone Marrow Transplant . 1990; 5 407-411
- 150 Attal M, Huguet F, Rubie H. Prevention of hepatic veno-occlusive disease after bone marrow transplantation by continuous infusion of low-dose heparin: a prospective, randomized trial. Blood . 1992; 79 1-7
- 151 Rosenthal J, Sender L, Secola R. Phase II trial of heparin prophylaxis for veno-occlusive disease of the liver in children undergoing bone marrow transplantation. Bone Marrow Transplant . 1996; 18 185-191
- 152 Marsa-Vila L, Gorin N C, Laporte J P. Prophylactic heparin does not prevent liver venocclusive disease following autologous bone marrow transplantation. Eur J Haematol . 1991; 47 346-354
- 153 Cahn J Y, Flesch M, Brion A. Prevention of veno-occlusive disease of the liver after bone marrow transplantation: heparin or no heparin?. Blood . 1992; 80 2149-2150
- 154 Or R, Nagler A, Shpilberg O. Low molecular weight heparin for the prevention of veno-occlusive disease of the liver in bone marrow transplantation patients. Transplantation . 1996; 61 1067-1071
- 155 Essell J H, Schroeder M T, Harman G S. Ursodiol prophylaxis against hepatic complications of allogeneic bone marrow transplantation. A randomized, double-blind, placebo-controlled trial. Ann Intern Med . 1998; 128 975-981
- 156 Ohashi K, Tanabe J, Watanabe R. The Japanese multicenter open randomized trial of ursodeoxycholic acid prophylaxis for hepatic veno-occlusive disease after stem cell transplantation. Am J Hematol . 2000; 64 32-38
- 157 Ruutu T, Eriksson B, Remes K. Ursodiol for the prevention of hepatic complications in allogeneic stem cell transplantation. Bone Marrow Transplant . 1999; 23 756(Abst)
- 158 Gluckman E, Jolivet I, Scrobohaci M L. Use of prostaglandin E1 for prevention of liver veno-occlusive disease in leukaemic patients treated by allogeneic bone marrow transplantation. Br J Haematol . 1990; 74 277-281
- 159 Bearman S I, Shen D, Hinds M S. A phase I/II study of prostaglandin E1 for the prevention of hepatic venocclusive disease after bone marrow transplantation. Br J Haematol . 1993; 84 724-730
- 160 Clift R A, Bianco J A, Appelbaum F R. A randomized controlled trial of pentoxifylline for the prevention of regimen-related toxicities in patients undergoing allogeneic marrow transplantation. Blood . 1993; 82 2025-2030
- 161 Attal M, Huguet F, Rubie H. Prevention of regimen-related toxicities after bone marrow transplantation by pentoxifylline: a prospective randomized trial. Blood . 1993; 82 732-736
- 162 Rubenfeld G D, Crawford S W. Withdrawing life support from mechanically ventilated recipients of bone marrow transplants: a case for evidence-based guidelines. Ann Intern Med . 1996; 125 625-633
- 163 Bearman S I, Shuhart M C, Hinds M S. Recombinant human tissue plasminogen activator for the treatment of established severe hepatic venocclusive disease of the liver after bone marrow transplantation. Blood . 1992; 80 2458-2462
- 164 Ringden O, Wennberg L, Ericzon B G. Alteplase for hepatic veno-occlusive disease after bone marrow transplantation [Letter]. Lancet . 1992; 340 546-547
- 165 Hagglund H, Ringden O, Ericzon B G. Treatment of hepatic venoocclusive disease with recombinant human tissue plasminogen activator or orthotopic liver transplantation after allogeneic bone marrow transplantation. Transplantation . 1996; 62 1076-1080
- 166 Palmer K J, Goa K L. Defibrotide. A review of its pharmacodynamic and pharmacokinetic properties, and therapeutic use in vascular disorders. Drugs . 1993; 45 259-294
- 167 Richardson P G, Elias A D, Krishnan A. Treatment of severe veno-occlusive disease with defibrotide: compassionate use results in efficacy without significant toxicity in a high risk population. Blood . 1998; 92 737-744
- 168 Chopra R, Eaton J D, Grassi A. Defibrotide for the treatment of hepatic veno-occlusive disease: results of the European compassionate-use study. Br J Haematol . 2000; 111 1122-1129
- 169 Haire W D, Ruby E I, Stephens L C. A prospective randomized double-blind trial of antithrombin III concentrate in the treatment of multiple-organ dysfunction syndrome during hematopoietic stem cell transplantation. Biol Blood Marrow Transplant . 1998; 4 142-150
- 170 Mertens R, Brost H, Granzen B. Antithrombin treatment of severe hepatic veno-occlusive disease in children with cancer. Eur J Pediatr . 1999; 158(Suppl 3) 154-158
- 171 Ibrahim A, Pico J L, Maraninchi D. Hepatic venocclusive disease following bone marrow transplantation treated by prostaglandin E1 . Bone Marrow Transplant . 1991; 7(Suppl 2) 53
- 172 Khoury H, Adkins D, Brown R. Does early treatment with high-dose methylprednisolone alter the course of hepatic regimen-related toxicity?. Bone Marrow Transplant . 2000; 25 737-743
- 173 Kajiume T, Yoshimi S, Nagita A. Application of nitric oxide for a case of veno-occlusive disease after peripheral blood stem cell transplantation. Paediatr Hematol Oncol . 2000; 17 601-604
- 174 Nattakom T V, Charlton A, Wilmore D W. Use of vitamin E and glutamine in the successful treatment of severe veno-occlusive disease following bone marrow transplantation. Nutr Clin Pract . 1995; 10 16-18
- 175 Smith F O, Johnson M S, Scherer L R. Transjugular intrahepatic portosystemic shunting (TIPS) for treatment of severe hepatic veno-occlusive disease. Bone Marrow Transplant . 1996; 18 643-646
- 176 Fried M W, Connaghan D G, Sharma S. Transjugular intrahepatic portosystemic shunt for the management of severe venoocclusive disease following bone marrow transplantation. Hepatology . 1996; 24 588-591
- 177 Azoulay D, Castaing D, Lemoine A. Transjugular intrahepatic portosystemic shunts (TIPS) for severe veno-occlusive disease of the liver following bone marrow transplantation. Bone Marrow Transplant . 2000; 25 987-992
- 178 Zenz T, Rossle M, Bertz H. Severe veno-occlusive disease after allogeneic bone marrow or peripheral stem cell transplantation-role of transjugular intrahepatic portosystemic shunt (TIPS). Liver . 2001; 21 31-36
- 179 Rajvanshi P, McDonald G B. Expanding the use of transjugular intrahepatic portosystemic shunts for veno-occlusive disease. Liver Transplant . 2001; 7 154-159
- 180 Meacher R, Venkatesh B, Lipman J. Acute respiratory distress syndrome precipitated by transjugular intrahepatic porto-systemic shunting for severe hepatic veno-occlusive disease. Is it due to pulmonary leucostasis?. Intensive Care Med . 1999; 25 1332-1333
- 181 Murray J A, LaBrecque D R, Gingrich R D. Successful treatment of hepatic venocclusive disease in a bone marrow transplant patient with side-to-side portacaval shunt. Gastroenterology . 1987; 92 1073-1077
- 182 Jacobson B K, Kalayoglu M. Effective early treatment of hepatic venocclusive disease with a central splenorenal shunt in an infant. J Pediatr Surg . 1992; 27 531-533
- 183 Nimer S D, Milewicz A L, Champlin R E. Successful treatment of hepatic venocclusive disease in a bone marrow transplant patient with orthotopic liver transplantation. Transplantation . 1990; 49 819-821
- 184 Rapoport A P, Doyle H R, Starzl T. Orthotopic liver transplantation for life-threatening veno-occlusive disease of the liver after allogeneic bone marrow transplant. Bone Marrow Transplant . 1991; 8 421-424
- 185 Salat C, Holler E, Wolf C. Laboratory markers of veno-occlusive disease in the course of bone marrow and subsequent liver transplantation. Bone Marrow Transplant . 1997; 19 487-490
- 186 Schlitt H J, Tischler H J, Ringe B. Allogeneic liver transplantation for hepatic veno-occlusive disease after bone marrow transplantation-clinical and immunological considerations. Bone Marrow Transplant . 1995; 16 473-478
- 187 Dowlati A, Honore P, Damas P. Hepatic rejection after orthotopic liver transplantation for hepatic veno-occlusive disease or graft-versus-host disease following bone marrow transplantation. Transplantation . 1995; 60 106-109
- 188 Bunin N, Leahey A, Dunn S. Related donor liver transplant for veno-occlusive disease following T-depleted unrelated donor bone marrow transplantation. Transplantation . 1996; 61 664-666
- 189 Rosen H R, Martin P, Schiller G J. Orthotopic liver transplantation for bone-marrow transplant-associated veno-occlusive disease and graft-versus-host disease of the liver. Liver Transplant Surg . 1996; 2 225-232

Figure 1 Photomicrographs illustrating the evolution of histologic abnormalities of SOS. (A and B) Fatal SOS 23 days postautograft (hematoxylin and eosin stain). (A) Hemorrhagic necrosis of zone 3 with sparing of zone 1 (PV, portal vein). (B) High-power methacrylate thin section showing early changes of concentric venular luminal narrowing by a widened subendothelial zone (double arrow) containing entrapped red cells. The surrounding changes include anuclear clusters of necrotic hepatocytes, disrupted sinusoids, and hemorrhage into the space of Disse. (C) Severe SOS 21 days postallograft (Gomori trichrome stain). Hepatocyte dropout, sinusoidal hemorrhage, and embolization of a cluster of hepatocytes (arrow) into a partially collagenized venule. (D) Immunostained liver section of early SOS with anti-Factor VIII/von Willebrand factor, showing intense perivenular and adventitial staining, loss of the hepatocyte background staining in zone 3, and preservation of portal and zone 1 anatomy. A similar perivenular pattern of deposition was seen with immunofluorescent anti-fibrogen immunostaining, whereas anti-platelet antigen immunostaining was negative.[81] (E) Fatal SOS 31 days postallograft (Mallory trichrome stain). A small sublobular venule is nearly occluded by loose extracellular matrix and red cells within the widened subendothelial zone (double arrows). Extensive necrosis has obliterated the liver cords, which are replaced by fine curvilinear strands of connective tissue admixed with red cells. (F and G) Fatal sinusoidal obstruction syndrome 37 days after (gemtuzumab ozogamicin infusion for relapsed acute myelogenous leukemia posttransplantation. (F) Extensive zone 3 sinusoidal fibrosis adjacent to a nonoccluded sublobular venule (Mallory trichrome stain). (G) Immunostain of the same specimen with anti-α smooth muscle actin showing proliferation of activated hepatic stellate cells lining perivenular sinusoids. (H) Fatal SOS 67 days postallograft (Mallory trichrome stain), showing a pattern of ``reverse'' cirrhosis with confluent bridging between collapsed, fibrotic perivenular zones containing venules occluded by fibrous matrix.
