4,647
Views
4
CrossRef citations to date
0
Altmetric
Drug Profile

Defibrotide sodium for the treatment of hepatic veno-occlusive disease/sinusoidal obstruction syndrome

, , , , , & show all
Pages 113-124 | Received 11 Aug 2017, Accepted 22 Dec 2017, Published online: 05 Jan 2018

ABSTRACT

Introduction: Hepatic veno-occlusive disease/sinusoidal obstruction syndrome (VOD/SOS) is an unpredictable condition associated with endothelial-cell damage due to conditioning for hematopoietic stem-cell transplantation (HSCT) or chemotherapy without HSCT. Mortality in patients with VOD/SOS and multi-organ dysfunction (MOD) may be >80%.

Areas covered: Defibrotide is the only approved drug for the treatment of severe hepatic VOD/SOS after HSCT in the European Union and hepatic VOD/SOS with renal or pulmonary dysfunction in the United States. Its efficacy in patients with VOD/SOS with MOD post-HSCT was demonstrated in a clinical-trial program that included a historically controlled treatment study, a phase 2 trial, and a large T-IND expanded-access program that also included patients without MOD and who received chemotherapy without HSCT.

Expert commentary: Defibrotide appears to protect endothelial cells and restore the thrombolytic–fibrinolytic balance. It addresses a significant clinical need and has demonstrated favorable Day +100 survival and overall adverse-event rates that seem similar to control groups receiving supportive care alone. Currently, defibrotide is under investigation for the prevention of VOD/SOS in high-risk pediatric and adult patients.

1. Introduction

Hepatic veno-occlusive disease (VOD), or sinusoidal obstruction syndrome (SOS), is an unpredictable, potentially life-threatening complication of conditioning regimens for hematopoietic stem cell transplantation (HSCT) [Citation1Citation3]. It is characterized by painful hepatomegaly, hyperbilirubinemia, rapid weight gain, and accumulation of ascitic fluid within the abdomen [Citation1]. VOD/SOS typically develops during the first 21 days following transplantation [Citation4], although delayed onset may occur [Citation3,Citation5,Citation6]. The mean incidence is estimated to be 13.7% (range 0–62%) following myeloablative conditioning for HSCT [Citation7,Citation8], and approximately 9% in a study of patients receiving HSCT with reduced-intensity conditioning [Citation9]. The mortality risk for patients who develop hepatic VOD/SOS with multi-organ dysfunction (MOD), also known as multi-organ failure, typically characterized by pulmonary and/or renal dysfunction, has been estimated to be >80% [Citation7].

VOD/SOS is not a disease restricted to the transplantation setting. It may also develop in patients receiving chemotherapy or calicheamicin–antibody drug conjugates not associated with HSCT [Citation1,Citation3,Citation10]. In a recent interim report from a large prospective treatment study of patients with VOD/SOS, approximately 11% of enrolled patients had not received HSCT [Citation11]. The incidence of VOD/SOS in acute lymphoblastic leukemia patients treated with inotuzumab ozogamicin is higher than in patients receiving standard chemotherapy (13% vs. <1%, respectively) [Citation10]. An increased risk for development of VOD/SOS, with or without HSCT, also has been well recognized in acute myeloid leukemia patients treated with another monoclonal antibody, gemtuzumab ozogamicin [Citation12].

In the transplantation setting, exposure to inotuzumab or gemtuzumab prior to HSCT increases the risk of developing VOD/SOS, which appears to be associated with a shorter time between drug treatment and the start of HSCT conditioning, as well as the type of conditioning [Citation10,Citation12]. Among 212 patients who received inotuzumab in 3 studies, VOD/SOS developed in 2 patients with prior HSCT, 14 with HSCT following inotuzumab, and 5 with HSCT both before and after inotuzumab treatment (of note, VOD/SOS also developed in 5 patients without HSCT) [Citation10]. In the retrospective study of gemtuzumab, 9 (64%) of 14 patients who received gemtuzumab prior to HSCT developed VOD/SOS, in contrast with 4 (8%) of 48 without gemtuzumab exposure prior to HSCT [Citation12].

The British Committee for Standards in Haematology (BCSH) and the British Society for Blood and Marrow Transplantation (BSBMT) guidelines on treatment of VOD/SOS recommend that all patients should be assessed for risk factors before HSCT [Citation4]. Among the known risk factors are pre-transplant patient characteristics (e.g. age, disease type, preexisting liver condition, prior treatment with certain chemotherapeutic agents, prior abdominal radiation) and transplant-related risk factors (e.g. graft type, conditioning regimen, graft-versus-host disease [GVHD] prophylaxis regimen) [Citation3,Citation13]. The BCSH/BSBMT further recommend that reversible patient risk factors (e.g. iron overload in patients with thalassemia) be appropriately addressed prior to HSCT [Citation4].

The multifactorial and complex pathophysiology of VOD/SOS is believed to involve a cascade of injury leading to sinusoidal endothelial cell (EC) and hepatocyte damage [Citation1Citation3]. It is believed that under conditions where metabolism is impaired (e.g. glutathione store depletion, drug–drug interaction leading to P450 inhibition), toxic chemotherapy and/or its metabolites accumulate in zone 3 of the hepatic acinus. Accumulation of these toxins leads to direct injury and ballooning of sinusoidal ECs, thus compromising the fenestrated endothelium and allowing extravasation of red blood cells into the space of Disse. The resulting extraluminal compression of the sinusoidal tract, combined with sinusoidal thrombosis triggered by platelet activation and proinflammatory/procoagulant activation from direct EC injury, ultimately leads to central venous and/or sinusoidal obstruction [Citation1,Citation2,Citation14,Citation15].

The sloughing of necrotic ECs from the endothelial lining further obstructs the sinusoidal flow [Citation2]. The result is obstruction of central venous blood flow and ischemia in zone 3. Later effects can include collagen deposition and fibrotic obstruction of the central and small hepatic veins [Citation15]. Sinusoidal obstruction may then lead to portal hypertension, reversal of hepatic venous flow, hepatorenal pathophysiology, and MOD [Citation1,Citation16,Citation17].

1.1. Diagnosis

Traditionally, VOD/SOS is diagnosed within 21 days of HSCT by the Baltimore criteria, which require hyperbilirubinemia, or within 20 days by the modified Seattle criteria, for which elevated bilirubin is not mandated [Citation17,Citation18] (). However, a wide range of presentations has been characterized in the past 20–30 years that is not covered by these criteria [Citation3], and important differences between adults and pediatric patients have been observed [Citation3,Citation19] (). Use of abdominal ultrasound with Dopplers to assess for portal venous blood flow reversal also may aid in diagnosis in cases where standard clinical criteria are not met. Finally, a liver biopsy, preferably a transjugular approach with measurement of transhepatic venous pressure gradients, can be extremely helpful to establish the diagnosis of VOD/SOS, and rule out other causes of liver injury such as hepatic GVHD [Citation3,Citation5].

Table 1. Criteria established for diagnosis of VOD/SOS patients [Citation5,Citation6,Citation18,Citation19].

Table 2. Major differences in the EBMT’s recently proposed diagnostic criteria for VOD/SOS [Citation6].

The European Society for Blood and Marrow Transplantation (EBMT) has recently proposed updated criteria for VOD/SOS [Citation5,Citation6] (), as well as detailed prospective severity grades that may facilitate earlier recognition of severe VOD/SOS in pediatric and adult patients [Citation5,Citation6], and guide appropriate treatment [Citation20] ().

Table 3. New EBMT criteria for severity grading of suspected VOD/SOS [Citation5,Citation6].

2. Body of review

2.1. Overview of treatments

Previous standard-of-care treatment strategies for VOD/SOS after HSCT have focused on supportive care measures, including adequate fluid and sodium balance, and avoidance of hepato- and nephrotoxic drugs [Citation3,Citation4,Citation20,Citation21], whereas an effective therapeutic treatment had remained an unmet medical need. However, clinical trials have now demonstrated the efficacy of defibrotide for the treatment of VOD/SOS in patients with MOD following HSCT, and it is today the only approved treatment available for these patients in the United States and the European Union (see Section 2.5) [Citation22,Citation23].

The clinical evidence for the efficacy and safety of alternative treatments to defibrotide is limited [Citation4]. Data are limited for methylprednisolone [Citation4,Citation24Citation26] and include a retrospective study of 9 pediatric patients (4 also treated with defibrotide), 8 with MOD, 7 of whom survived [Citation25], and the BCSH/BSBMT guideline notes that it may be considered for use in the treatment of VOD/SOS although caution is recommended due to the risk of infections [Citation4]. On the other hand, N-acetylcysteine did not improve liver toxicity after HSCT [Citation27] and is not routinely recommended for use in the treatment of VOD/SOS due to lack of efficacy [Citation4,Citation27,Citation28]. Fibrinolytic treatment strategies using tissue plasminogen activator (tPA), with or without heparin, have been assessed in a number of small studies [Citation4]. One of the larger studies of tPA (n = 42) retrospectively found 25% Day +100 survival but a 24% rate of severe hemorrhage [Citation29]. Another retrospective study (n = 56) did not report Day +100 survival but suggested that dose-limited tPA may reduce the risk of hemorrhage [Citation30]. Fibrinolytic treatments are generally not recommended by BCSH/BSBMT due to the associated risk of hemorrhage [Citation4]. The BCSH/BSBMT guideline does not recommend prostaglandin E1, pentoxifylline, heparin (unfractionated and low molecular weight), or antithrombin as prophylaxis due to lack of efficacy and/or increased toxicity [Citation4,Citation31]. Treatment with ursodeoxycholic acid can reduce hydrophobic bile acids that can be toxic to hepatic parenchymal cells, which may reduce the incidence of severe acute GVHD in HSCT patients, and it has been suggested as prophylaxis for VOD/SOS [Citation3,Citation4,Citation32,Citation33]; however, clinical results have been mixed, with a systematic review showing a relative risk of VOD/SOS of 34–36% in adults compared with no treatment [Citation32] while other studies do not, including a prospective, controlled study in 242 HSCT patients that found no difference in incidence of VOD/SOS [Citation33]. In certain cases of refractory ascites and severe portal hypertension, a transjugular intrahepatic portosystemic shunt, or even hepatic transplantation, may be considered after consultation with hepatology and critical care specialists [Citation4,Citation34Citation37].

2.2. Introduction to defibrotide sodium

2.2.1. Chemistry

Defibrotide (sodium salt) is a complex polydisperse mixture of primarily single-stranded oligodeoxyribonucleotides derived from mammalian tissue (porcine mucosa) by controlled depolymerization [Citation38Citation42]. It has aptameric activity on the vascular endothelium and antithrombotic, thrombolytic, anti-inflammatory, and anti-ischemic properties [Citation43Citation45].

2.2.2. Pharmacodynamics

In vitro, defibrotide has been shown to bind to various sites predominantly located on the plasma membranes of the vascular endothelium [Citation44,Citation46]. It acts as an adenosine receptor agonist and is thought to have affinity for receptors A1 and A2 [Citation39], although a recent in vitro study was unable to find support for this interaction [Citation46].

Additional in vitro data show that defibrotide is internalized into the ECs, where it promotes protection of activated ECs through profibrinolytic and antithrombotic effects, as well as exhibiting anti-inflammatory and anti-adhesion activity [Citation22,Citation23,Citation44,Citation46,Citation47]. Defibrotide may also provide mild anticoagulant protection of ECs from damage caused by chemotherapy, tumor necrosis factor α, serum starvation, and perfusion, and it may decrease influx and activity of inflammatory mediators, including leukocyte adhesion molecules, without affecting the clinical efficacy of chemotherapy [Citation47Citation50]. Additionally, defibrotide has been shown to attenuate the generation of reactive oxygen species and restore endothelial nitric oxide synthase levels in the face of oxidative stress [Citation46].

Through increased tPA and thrombomodulin expression, decreased von Willebrand factor, decreased heparanase expression, and decreased plasminogen activator inhibitor-1 activity, defibrotide decreases the procoagulant activity and increases the enzymatic, fibrinolytic activity of plasmin, which support extracellular matrix integrity, tissue homeostasis, and EC structure, as shown in vitro [Citation22,Citation23,Citation39,Citation40,Citation47,Citation48,Citation51Citation53].

Defibrotide (originally called ‘Fraction P’) was initially approved for the treatment of thrombophlebitis and as prophylaxis of deep vein thrombosis in Italy [Citation44,Citation54]. The rationale for the first use in VOD/SOS began with a search for an agent with demonstrated antithrombotic properties that did not seem to be associated with a substantial increase in bleeding risk [Citation55]. A number of studies suggested the potential efficacy of defibrotide in vascular disorders, including peripheral vascular disease, microvascular thrombotic states, and chemotherapy-related hemolytic uremic syndrome [Citation54,Citation56Citation58]. Defibrotide has been demonstrated to have no significant or clinically relevant effect on cardiac electrophysiology with no prolongation of the QTc interval at doses 2.4–4 times higher than therapeutically indicated [Citation22,Citation23]. Tolerability of defibrotide appeared to be acceptable with a relative lack of systemic anticoagulant activity, which suggested a possible therapeutic advantage over other available treatments, especially when coupled with a potential for ameliorating diverse manifestations of vascular injury [Citation55]. Based on these properties, however, concomitant use of thrombolytic therapy (e.g. tPA) is contraindicated [Citation22,Citation23].

2.2.3. Pharmacokinetics and metabolism

Defibrotide is administered in 4 doses per day (every 6 h) of 6.25 mg/kg (i.e. 25 mg/kg/day) given as 2-h infusions [Citation22,Citation23]. In healthy subjects, defibrotide has a half-life of ~43 min, Tmax at 2 h (end of infusion), and does not induce or inhibit cytochrome P450 metabolism [Citation22,Citation23]. An open-label phase 1 study on the effect of hemodialysis and on severe or end-stage renal disease on defibrotide pharmacokinetics found that dose adjustments in patients with renal impairment are not required due to limited urinary excretion and lack of accumulation [Citation59].

2.3. Clinical efficacy

2.3.1. Early phase/phase 1 studies

The clinical safety, tolerability, and efficacy of defibrotide in VOD/SOS with MOD were first evaluated in a multicenter emergency use program in 1998 [Citation55]. Other early trials provided consistent, promising results, including the early European compassionate-use study [Citation60], and an expansion of the initial study [Citation61]. A retrospective multicenter study in pediatric patients with and without MOD found that Day +100 survival was 64% after treatment with defibrotide, with a corresponding overall rate of complete response to treatment (i.e. resolution of symptoms of VOD/SOS and MOD) of 76% [Citation45] (). In general, phase 1 studies reported complete response rates of 36–76% and Day +100 survival rates of 32–64% [Citation45,Citation51,Citation55,Citation60,Citation61].

Table 4. Key early phase clinical studies of defibrotide for treatment of VOD/SOS [Citation62].

2.3.2. Phase 2 studies

An open-label, prospective, multicenter, randomized dose-finding phase 2 trial compared 2 doses of defibrotide (25 mg/kg/day [n = 75] and 40 mg/kg/day [n = 74]) and found no significant difference in efficacy between doses [Citation51]. In the study, the minimum recommended duration of treatment was 14 days; however, defibrotide was administered for medians of 19 and 20 days, respectively, in the 25- and 40-mg/kg/day arms. Rates of complete response, the primary endpoint, and Day +100 survival post-HSCT were 46% and 42%, respectively. The overall incidence of treatment-related adverse events (AEs) was similar for the low and high doses (7% and 10%, respectively). Pediatric patients demonstrated similar incidences of bleeding events for the low and high doses (68% vs. 52%; = 0.377), although hypotension was apparently more common among those receiving the higher dose (52% vs. 26%; = 0.083). Those receiving the higher dose had a numerically higher incidence of AEs assessed as being likely or definitely related to defibrotide (68% vs. 39%; = 0.081) [Citation51]. Based on these efficacy and safety findings, defibrotide 25 mg/kg/day was selected as the dosage for investigation in the historically controlled treatment trial discussed in Section 2.3.3, with a recommended minimum duration of 21 days.

In a large international multicenter compassionate use program (n = 710), conducted between December 1998 and March 2009 [Citation63], defibrotide at 10, 25, 40, 60, or 80 mg/kg/day was administered for a median of 15 days to patients with VOD/SOS, with or without MOD (89% of patients had received HSCT). While the overall Kaplan–Meier estimated Day +100 survival was 54%, it was 58% for the 25 mg/kg/day dose group (the approved dose; 65.4% in the pediatric and 46.1% in the adult 25 mg/kg/day dose subgroups). Patients without MOD had an estimated survival of 64.7% compared with 39.7% among those with MOD. The highest estimated survival, 74.2%, was in the subgroup of non-HSCT patients without MOD (n = 41). AEs were reported by 53% of patients, most commonly MOD (20%), progression of VOD/SOS (11%), sepsis (7%), and GVHD (4%). In the 25 mg/kg/day dose-group, AEs were reported by 47% of patients. Overall safety and efficacy were consistent with prior studies on defibrotide in VOD/SOS ( and ) [Citation62,Citation63].

Figure 1. Outcome from international compassionate-use program [Citation63]. Survival to Day +100 by dose (N = 701). a

Vertical lines signify censored data. aExcludes nine patients with missing HSCT or nontransplantation-associated chemo-radiotherapy dates. bApproved dose in the European Union and in the United States. CI, confidence interval; HSCT, hematopoietic stem cell transplantation. Reprinted from Biology of Blood and Marrow Transplantation, 22/10, Corbacioglu S, et al, Defibrotide for the Treatment of Hepatic Veno-Occlusive Disease: Final Results From the International Compassionate-Use Program, 1874–1882, Copyright (2016), with permission from American Society for Blood and Marrow Transplantation. doi:10.1016/j.bbmt.2016.07.001. URL for Creative Commons user license: https://creativecommons.org/licenses/by-nc-nd/4.0/.

Figure 1. Outcome from international compassionate-use program [Citation63]. Survival to Day +100 by dose (N = 701). aVertical lines signify censored data. aExcludes nine patients with missing HSCT or nontransplantation-associated chemo-radiotherapy dates. bApproved dose in the European Union and in the United States. CI, confidence interval; HSCT, hematopoietic stem cell transplantation. Reprinted from Biology of Blood and Marrow Transplantation, 22/10, Corbacioglu S, et al, Defibrotide for the Treatment of Hepatic Veno-Occlusive Disease: Final Results From the International Compassionate-Use Program, 1874–1882, Copyright (2016), with permission from American Society for Blood and Marrow Transplantation. doi:10.1016/j.bbmt.2016.07.001. URL for Creative Commons user license: https://creativecommons.org/licenses/by-nc-nd/4.0/.

2.3.3. Comparative studies of defibrotide for VOD/SOS treatment

In a historically controlled treatment trial, the efficacy and safety of defibrotide at 25 mg/kg/day in VOD/SOS with MOD were compared with those of patients from a historical control group who met the study’s inclusion criteria but were treated at the study’s centers prior to the availability of defibrotide at that institution () [Citation64]. Initiation of a randomized study had been rejected on ethical grounds due to the life-threatening nature of VOD/SOS with MOD, the lack of effective alternative agents, and compelling evidence of efficacy from several prior defibrotide studies. To identify an appropriate control group, nearly 7000 medical charts of HSCT patients hospitalized at each participating center were sequentially reviewed in reverse chronological order starting 6 months prior to the first approved use of defibrotide at that site. This methodology was designed to control for institutional standards of care. Treatment was recommended for at least 21 days, and the median duration of treatment was 21 days, which was similar to the 21-day timeframe between median day of diagnosis and onset of complete response. The trial confirmed significantly higher Day +100 survival rates for patients receiving defibrotide vs. best supportive care (38.2% vs. 25.0%, respectively, with a propensity-adjusted between-group estimated difference of 23%; = 0.0109) and higher Day +100 complete response rates (25.5% vs. 12.5%, respectively, propensity-adjusted between-group estimated difference of 19%; = 0.160), with tolerability that was generally similar between arms. AEs occurred at a frequency of 98% and 100% for the defibrotide and historical control groups, respectively. The most common AEs were hypotension (39.2% and 50.0%, respectively) and diarrhea (23.5% and 37.5%, respectively). No difference was observed in the overall incidence of common hemorrhagic AEs (64% and 75%, respectively) [Citation64].

Figure 2. Kaplan–Meier estimated survival to Day +100 in the historically controlled treatment trial [Citation64].

Republished with permission of American Society of Hematology, from Phase 3 Trial of Defibrotide for the Treatment of Severe Veno-Occlusive Disease and Multi-Organ Failure, Richardson PG, et al, 127, 13 and 2016; permission conveyed through Copyright Clearance Center, Inc.

Figure 2. Kaplan–Meier estimated survival to Day +100 in the historically controlled treatment trial [Citation64].Republished with permission of American Society of Hematology, from Phase 3 Trial of Defibrotide for the Treatment of Severe Veno-Occlusive Disease and Multi-Organ Failure, Richardson PG, et al, 127, 13 and 2016; permission conveyed through Copyright Clearance Center, Inc.

An expanded-access treatment (T-IND) protocol for the treatment of VOD/SOS in patients with and without MOD, had the clinical objective to make defibrotide 25 mg/kg/day available for patients and generate prospective data on outcomes [Citation11]. This open-label, single-arm study was designed to provide access to defibrotide and did not include onsite monitoring. Interim results from the T-IND study, which encompassed a broadly inclusive VOD/SOS patient population and was the largest prospective evaluation of defibrotide for treatment of VOD/SOS to date (N = 642), showed an overall Day +100 survival of 50.3% and 45.3% for the subgroup with MOD. The Day +100 survival was 54.5% and 44.9% in the pediatric and adult subgroups, respectively. AEs were reported in 69.6% of patients, most commonly hypotension (13.8%) and new or worsening MOD (12.7%). Overall, AEs assessed as treatment-related were reported in 21.6% of patients, and were generally similar between patients with and without MOD. The most common treatment-related AEs were pulmonary hemorrhage (4.7%), gastrointestinal hemorrhage (3.3%), and epistaxis (3.1%), which were consistent with prior studies of defibrotide [Citation54]. The recommended duration of treatment was ≥21 days (median, 21 days) [Citation11], which was similar to the previously observed length between median time to diagnosis (13 days) and median time to onset of complete response (34.5 days) in the historically controlled treatment study [Citation64], as well as the median length of treatment in a phase 2 trial [Citation51].

A post-hoc exploratory analysis of 573 HSCT patients from the T-IND study, including 351 (61.3%) with MOD, demonstrated a significant trend for improved Day +100 survival with earlier treatment initiation post-diagnosis, which was confirmed by the Cochran-Armitage trend test (< 0.001) in both the overall post-HSCT population and the MOD subgroup [Citation65]. Overall, 31.9% of patients received defibrotide on the day of diagnosis, and 93.0% had initiated treatment by day 7 post-diagnosis; reason for treatment delay was not assessed. Moreover, there was no day post-diagnosis that provided a clinically apparent cutoff for better outcomes, and earlier treatment was consistently associated with a survival benefit. These results are consistent with a previous pediatric study, which reported that patients with complete response had an average treatment delay of 1 day from diagnosis to initiation of treatment, compared with an average 5.5 days in those who did not achieve complete response [Citation45].

The results from the historically controlled treatment trial are further supported by those of a study using retrospective data from the Center for International Blood and Marrow Transplantation Research (CIBMTR); 8341 patients who underwent allogeneic HSCT between 2008 and 2011 were retrieved; 3.2% met criteria for VOD/SOS, and 1.2% met criteria for VOD/SOS with MOD [Citation66]. Among these patients with VOD/SOS, 1.2% (n = 101) had concomitant MOD and 3.3% (n = 275) did not have MOD. For analysis, patients with VOD/SOS and MOD were further divided into two groups: treated (n = 41) and not treated (n = 55) with defibrotide. The overall Day +100 survival was 39% in patients receiving defibrotide compared with 30.9% in those not receiving defibrotide. The Day +100 survival was 40.0% and 37.5% for pediatric and adult patients receiving defibrotide, respectively. Resolution of VOD/SOS at Day +100 was 51% in the defibrotide group (56% for pediatric and 43.8% for adult patients), and 29% in the non-defibrotide group. In addition, defibrotide was associated with reduced incidence (23.1% vs. 37.7%) and severity of acute GVHD compared with patients not receiving defibrotide () [Citation66].

Table 5. Comparative studies of defibrotide for VOD/SOS treatment [Citation62].

2.3.4. Phase 3 studies for the prevention of VOD/SOS

With the goal of assessing the prophylactic effects of defibrotide on the development of VOD/SOS, an open-label, randomized, controlled phase 3 trial of 356 pediatric patients at high risk of developing VOD/SOS post-HSCT was conducted [Citation41]. Patients were randomly allocated to receive defibrotide at 25 mg/kg/day for VOD/SOS prevention vs best supportive care. Patients in either arm were allowed to receive defibrotide as treatment if VOD/SOS developed. The study reported a substantial reduction in VOD/SOS onset by Day +30 post-HSCT for the defibrotide prophylaxis group vs control group (incidence 12% vs 20%; = 0.0488, Z test; = 0.0507, log-rank test). The AE rates were similar between groups. Moreover, as in the CIBMTR observational study, the rate of acute GVHD was reduced in the defibrotide arm at 30 (= 0.0057) and 100 days (= 0.0034) [Citation41].

Defibrotide is being investigated in an ongoing international trial as prophylaxis in adult and pediatric patients receiving HSCT who are at high and very-high-risk of developing VOD/SOS (ClinicalTrials.gov identifier, NCT02851407) [Citation67].

2.3.5. Safety profile and tolerability

The incidence of AEs in patients receiving defibrotide is comparable to that in control groups, which suggests that the observed AEs may be associated with preexisting illnesses, comorbidities, or complications of the HSCT, rather than the treatment [Citation41,Citation54,Citation64]. Among the AEs assessed as treatment-related, hemorrhage and hypotension were found to be the most common [Citation41,Citation51,Citation54,Citation64]. AE reporting methodology differed for individual studies; however, they were generally consistent with the safety profile found in the historically controlled trial [Citation64], which reported an overall AE incidence of 99% in the treated patients and 100% in the untreated controls. Hypotension was the most frequent AE (39% for defibrotide, 50% for controls), and common hemorrhagic AEs, which included pulmonary alveolar and gastrointestinal hemorrhage, occurred in 64% of defibrotide-treated patients and 75% of controls. Related AEs included hemorrhage and hypotension [Citation64]. In addition to VOD/SOS, defibrotide has demonstrated acceptable tolerability in the treatment of severe malaria, as well as a number of cardiovascular conditions, such as deep vein thrombosis, chronic venous insufficiency, peripheral vascular disorders, thrombophlebitis, microangiopathy, and acute myocardial infarction [Citation54].

2.4. Regulatory affairs

Originally approved in Italy for the treatment of thrombophlebitis and prevention of deep vein thrombosis, and later for the treatment of mild-to-moderate peripheral obstructive arteriopathy, defibrotide has been investigated for the treatment of peripheral obliterative arterial disease, arterial sufficiency, and symptoms of phlebitis [Citation68]. The drug was ultimately withdrawn in 2009 at the request of the marketing authorization holder [Citation69], primarily to support investigation of other indications.

In October 2013, defibrotide was granted marketing authorization by the European Commission (European Medicines Agency) as the first approved treatment of severe hepatic VOD/SOS after HSCT therapy, indicated in adults and pediatric patients ≥1 month of age [Citation69]. In March 2016, defibrotide became the first drug approved by the United States Food and Drug Administration for the treatment of adult and pediatric patients with hepatic VOD/SOS and renal or pulmonary dysfunction following HSCT [Citation22,Citation70].

3. Conclusions

Hepatic VOD/SOS is a potentially fatal condition that is thought to result from a pathophysiologic cascade following EC damage, frequently due to the conditioning regimen for HSCT [Citation3]. VOD/SOS with MOD has been associated with a mortality rate of >80% by Day +100 post-HSCT [Citation7]. Defibrotide has demonstrated efficacy in the treatment of these critically ill patients when compared with supportive care alone and has an overall AE rate that is similar to that of the best supportive care [Citation5,Citation54].

Defibrotide appears to protect the ECs at a key early juncture in the pathophysiologic cascade of VOD/SOS, through its profibrinolytic and antithrombotic effects, as well as its anti-inflammatory and anti-adhesion properties [Citation46,Citation54].

Defibrotide is the only approved drug in the United States and the European Union with VOD/SOS indications: 25 mg/kg/day for at least 21 days for the treatment of VOD/SOS post-HSCT [Citation3,Citation5,Citation6,Citation22,Citation23], in patients with severe VOD/SOS in the European Union and in patients with pulmonary or renal dysfunction in the United States (treatment up to 60 days). Moreover, earlier initiation of treatment after diagnosis has been associated with significantly improved Day +100 survival in a post-hoc analysis [Citation65]. Although defibrotide is still under active investigation for prevention of VOD/SOS in high-risk adult and pediatric patients scheduled to undergo HSCT, a previous phase 3 pediatric prevention trial showed promising results.

4. Expert commentary

Although developments in transplantation technique and evolution of supportive care have improved the survival for patients with VOD/SOS, defibrotide meets a significant current clinical need. Defibrotide, which is thought to restore the thrombolytic–fibrinolytic balance and provides EC protection, has consistently demonstrated favorable Day +100 survival rates while offering an AE rate that is similar to best supportive care. The rarity and dismal outcomes associated with VOD/SOS with MOD impose ethical limitations on study design; however, when all the data are taken together, these studies show a consistent effect across a large proportion of patients with this disease. In contrast, alternative and unapproved treatment options typically have limited support for efficacy and/or problematic safety profiles. Defibrotide is the only therapy approved in the United States for treatment of VOD/SOS with renal or pulmonary dysfunction post-HSCT and the European Union for the treatment of severe VOD/SOS post-HSCT.

Improved management and care of patients pre-HSCT, systematic risk assessment and management, rapid and accurate diagnosis, and prompt initiation of defibrotide treatment post-diagnosis are important tools to improve the Day +100 survival. Looking ahead, further research on defibrotide as prophylaxis for VOD/SOS is warranted, and an ongoing phase 3 trial is investigating the efficacy and safety of this drug plus best supportive care compared with best supportive care alone in the prevention of hepatic VOD/SOS in high- or very-high-risk adult and pediatric patients undergoing HSCT (ClinicalTrials.gov identifier NCT02851407).

5. Five-year view

Adoption of the new EBMT diagnosis and severity guidelines will help refine identification and treatment protocols for patients with VOD/SOS. More accurate delineation of risk factors for VOD/SOS could encompass identification of potential biomarkers and dynamic clinical markers that could alert clinicians to earlier diagnosis and subsequently, earlier initiation of treatment with defibrotide. In addition, results of the ongoing prophylaxis trial (NCT02851407) will inform decisions about the use of defibrotide to prevent occurrence of VOD/SOS. Finally, other areas of interest for further investigation could build on the current understanding of the therapeutic profile of defibrotide, and identify specific subgroups that may benefit from treatment, such as sirolimus-exposed patients, prior gemtuzumab or inotuzumab use, and patients with mismatched, unrelated allogeneic HSCT. These could include prophylaxis in specific patient cohorts (e.g. patients undergoing high-risk allogeneic HSCT; Clinicaltrials.gov identifier NCT02851407), combination therapies using defibrotide and other endothelial targeting agents, endothelial injury/activation and thrombofibrinolytic effects in conditions with similar underlying pathophysiology, the role in other syndromes characterized by microangiopathy [Citation54,Citation56], and effect on prevention of acute GVHD [Citation41,Citation71,Citation72].

Key issues

  • Hepatic veno-occlusive disease/sinusoidal obstruction syndrome (VOD/SOS) is associated with conditioning for hematopoietic stem-cell transplantation (HSCT), calicheamicin-antibody drug conjugates, or chemotherapy alone, which may damage the sinusoidal endothelium and lead to platelet activation and thrombosis. In patients with VOD/SOS and multi-organ dysfunction (MOD), the mortality rate may be >80%.

  • Defibrotide, which is thought to restore thrombofibrinolytic balance, is the only drug approved for the treatment of severe VOD/SOS with MOD in adult and pediatric patients after HSCT.

  • Efficacy of defibrotide was demonstrated in a historically controlled treatment study in patients with VOD/SOS and MOD and a phase 2 trial; these results were supported by those of a large phase 3 T-IND expanded-access study, which included subgroups (pediatric and adult patients, post-chemotherapy, VOD/SOS without MOD).

  • Safety of defibrotide seems to be similar to that of best supportive care alone. Rates of adverse events were similar between treatment and control populations (eg, a historically controlled treatment study and a phase 3 pediatric prevention study).

  • Defibrotide is not approved for use as prophylaxis; however, a phase 3 trial found reduced incidence of VOD/SOS in a high-risk pediatric population treated with prophylactic defibrotide, and the drug is being further investigated in an ongoing phase 3 trial in adults and children (ClinicalTrials.gov identifier, NCT02851407).

Declaration of interest

P Richardson has served on an advisory board and received research funding from Jazz Pharmaceuticals. V Ho has received honoraria from Jazz Pharmaceuticals. N Chao reports research funding from Jazz Pharmaceuticals. F Dignan has received honoraria and research funding from Jazz Pharmaceuticals. M Mohty has received honoraria and research funding from Jazz Pharmaceuticals. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed. Peer reviewers on this manuscript have no relevant financial or other relationships to disclose. Medical writing assistance supported by Jazz Pharmaceuticals was provided by Daniel Famer, PhD, The Curry Rockefeller Group and John Norwood, The Curry Rockefeller Group.

Additional information

Funding

Editorial support for this manuscript was funded by Jazz Pharmaceuticals.

References

  • Bearman SI. The syndrome of hepatic veno-occlusive disease after marrow transplantation. Blood. 1995;85(11):3005–3020.
  • Carreras E, Diaz-Ricart M. The role of the endothelium in the short-term complications of hematopoietic SCT. Bone Marrow Transplant. 2011;46(12):1495–1502.
  • Mohty M, Malard F, Abecassis M, et al. Sinusoidal obstruction syndrome/veno-occlusive disease: current situation and perspectives—a position statement from the European Society for Blood and Marrow Transplantation (EBMT). Bone Marrow Transplant. 2015;50(6):781–789.
  • Dignan FL, Wynn RF, Hadzic N, et al. BCSH/BSBMT guideline: diagnosis and management of veno-occlusive disease (sinusoidal obstruction syndrome) following haematopoietic stem cell transplantation. Br J Haematol. 2013;163(4):444–457.
  • Mohty M, Malard F, Abecassis M, et al. Revised diagnosis and severity criteria for sinusoidal obstruction syndrome/veno-occlusive disease in adult patients: a new classification from the European Society for Blood and Marrow Transplantation. Bone Marrow Transplant. 2016;51(7):906–912.
  • Corbacioglu S, Carreras E, Ansari M, et al. Diagnosis and severity criteria for sinusoidal obstruction syndrome/veno-occlusive disease in pediatric patients: a new classification from the European society for blood and marrow transplantation (EBMT) [published online July 31, 2017]. Bone Marrow Transplant. 2017. DOI:10.1038/bmt.2017.161.
  • Coppell JA, Richardson PG, Soiffer R, et al. Hepatic veno-occlusive disease following stem cell transplantation: incidence, clinical course, and outcome. Biol Blood Marrow Transplant. 2010;16(2):157–168.
  • Dvorak CC, Nejadnik B, Cao Z, et al. Incidence of hepatic veno-occlusive disease (VOD) in Premier research database (BMT Tandem abstract 421). Biol Blood Marrow Transplant. 2016;22(3 Suppl):S288.
  • Tsirigotis PD, Resnick IB, Avni B, et al. Incidence and risk factors for moderate-to-severe veno-occlusive disease of the liver after allogeneic stem cell transplantation using a reduced intensity conditioning regimen. Bone Marrow Transplant. 2014;49(11):1389–1392.
  • Kantarjian HM, DeAngelo DJ, Advani AS, et al. Hepatic adverse event profile of inotuzumab ozogamicin in adult patients with relapsed or refractory acute lymphoblastic leukaemia: results from the open-label, randomised, phase 3 INO-VATE study. Lancet Haematol. 2017;4(8):e387–e398.
  • Richardson PG, Smith AR, Triplett BM, et al. Defibrotide for patients with hepatic veno-occlusive disease/sinusoidal obstruction syndrome (VOD/SOS): interim results from a treatment IND study. Biol Blood Marrow Transplant. 2017;23(6):997–1004.
  • Wadleigh M, Richardson PG, Zahrieh D, et al. Prior gemtuzumab ozogamicin exposure significantly increases the risk of veno-occlusive disease in patients who undergo myeloablative allogeneic stem cell transplantation. Blood. 2003;102(5):1578–1582.
  • Dalle JH, Giralt SA. Hepatic veno-occlusive disease after hematopoietic stem cell transplantation: risk factors and stratification, prophylaxis, and treatment. Biol Blood Marrow Transplant. 2016;22(3):400–409.
  • Hunt BJ, Jurd KM. Endothelial cell activation. A central pathophysiological process. BMJ. 1998;316(7141):1328–1329.
  • Shulman H. Hepatic veno-occlusive disease after bone marrow transplantation. Immunohistochemical identification of the material within occluded central venules. Am J Pathol. 1987;127(3):549–558.
  • Bearman SI, Shuhart MC, Hinds MS, et al. Recombinant human tissue plasminogen activator for the treatment of established severe venocclusive disease of the liver after bone marrow transplantation. Blood. 1992;80(10):2458–2462.
  • McDonald GB, Hinds MS, Fisher LD, et al. Veno-occlusive disease of the liver and multiorgan failure after bone marrow transplantation: a cohort study of 355 patients. Ann Intern Med. 1993;118(4):255–267.
  • Jones RJ, Lee KS, Beschorner WE, et al. Venoocclusive disease of the liver following bone marrow transplantation. Transplantation. 1987;44(6):778–783.
  • Myers KC, Dandoy C, El-Bietar J, et al. Veno-occlusive disease of the liver in the absence of elevation in bilirubin in pediatric patients after hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2015;21(2):379–381.
  • Carreras E. How I manage sinusoidal obstruction syndrome after haematopoietic cell transplantation. Br J Haematol. 2015;168(4):481–491.
  • Richardson PG, Ho VT, Cutler C, et al. Hepatic veno-occlusive disease after hematopoietic stem cell transplantation: novel insights to pathogenesis, current status of treatment, and future directions. Biol Blood Marrow Transplant. 2013;19(1 Suppl):S88–S90.
  • Defitelio® (defibrotide sodium) [US PI]. Prescribing information (United States). Palo Alto (CA): Jazz Pharmaceuticals, Inc; 2016.
  • Defitelio® (defibrotide) [EU SPC]. Summary of product characteristics (Europe). Palo Alto (CA): Jazz Pharmaceuticals, Inc; 2016.
  • Khoury H, Adkins D, Brown R, et al. Does early treatment with high-dose methylprednisolone alter the course of hepatic regimen-related toxicity? Bone Marrow Transplant. 2000;25(7):737–743.
  • Myers KC, Lawrence J, Marsh RA, et al. High-dose methylprednisolone for veno-occlusive disease of the liver in pediatric hematopoietic stem cell transplantation recipients. Biol Blood Marrow Transplant. 2013;19(3):500–503.
  • Al Beihany A, Al Omar H, Sahovic E, et al. Successful treatment of hepatic veno-occlusive disease after myeloablative allogeneic hematopoietic stem cell transplantation by early administration of a short course of methylprednisolone. Bone Marrow Transplant. 2008;41(3):287–291.
  • Barkholt L, Remberger M, Hassan Z, et al. A prospective randomized study using N-acetyl-L-cysteine for early liver toxicity after allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant. 2008;41:785–790.
  • Ringden O, Remberger M, Lehmann S, et al. N-acetylcysteine for hepatic veno-occlusive disease after allogeneic stem cell transplantation. Bone Marrow Transplant. 2000;25(9):993–996.
  • Bearman SI, Lee JL, Barón AE, et al. Treatment of hepatic venocclusive disease with recombinant human tissue plasminogen activator and heparin in 42 marrow transplant patients. Blood. 1997;89(5):1501–1506.
  • Yoon JH, Min WS, Kim HJ, et al. Experiences of t-PA use in moderate-to-severe hepatic veno-occlusive disease after hematopoietic SCT: is it still reasonable to use t-PA? Bone Marrow Transplant. 2013;48(12):1562–1568.
  • Imran H, Tleyjeh IM, Zirakzadeh A, et al. Use of prophylactic anticoagulation and the risk of hepatic veno-occlusive disease in patients undergoing hematopoietic stem cell transplantation: a systematic review and meta-analysis. Bone Marrow Transplant. 2006;37(7):677–686.
  • Tay J, Tinmouth A, Fergusson D, et al. Systematic review of controlled clinical trials on the use of ursodeoxycholic acid for the prevention of hepatic veno-occlusive disease in hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2007;13(2):206–217.
  • Ruutu T, Eriksson B, Remes K, et al. Ursodeoxycholic acid for the prevention of hepatic complications in allogeneic stem cell transplantation. Blood. 2002;100(6):1977–1983.
  • Campos-Varela I, Castells L, Dopazo C, et al. Transjugular intrahepatic portosystemic shunt for the treatment of sinusoidal obstruction syndrome in a liver transplant recipient and review of the literature. Liver Transpl. 2012;18(2):201–205.
  • Smith FO, Johnson MS, Scherer LR, et al. Transjugular intrahepatic portosystemic shunting (TIPS) for treatment of severe hepatic veno-occlusive disease. Bone Marrow Transplant. 1996;18(3):643–646.
  • Zenz T, Rossle M, Bertz H, et al. Severe veno-occlusive disease after allogeneic bone marrow or peripheral stem cell transplantation–role of transjugular intrahepatic portosystemic shunt (TIPS). Liver. 2001;21(1):31–36.
  • Schoppmeyer K, Lange T, Wittekind C, et al. TIPS for veno-occlusive disease following stem cell transplantation. Z Gastroenterol. 2006;44(6):483–486.
  • Richardson P, Linden E, Revta C, et al. Use of defibrotide in the treatment and prevention of veno-occlusive disease. Expert Rev Hematol. 2009;2(4):365–376.
  • Bianchi G, Barone D, Lanzarotti E, et al. Defibrotide, a single-stranded polydeoxyribonucleotide acting as an adenosine receptor agonist. Eur J Pharmacol. 1993;238(2–3):327–334.
  • Echart CL, Graziadio B, Somaini S, et al. The fibrinolytic mechanism of defibrotide: effect of defibrotide on plasmin activity. Blood Coagul Fibrinolysis. 2009;20(8):627–634.
  • Corbacioglu S, Cesaro S, Faraci M, et al. Defibrotide for prophylaxis of hepatic veno-occlusive disease in paediatric haemopoietic stem-cell transplantation: an open-label, phase 3, randomised controlled trial. Lancet (London, England). 2012;379(9823):1301–1309.
  • Francischetti IM, Oliveira CJ, Ostera GR, et al. Defibrotide interferes with several steps of the coagulation-inflammation cycle and exhibits therapeutic potential to treat severe malaria. Arterioscler Thromb Vasc Biol. 2012;32(3):786–798.
  • Bracht F, Schror K. Isolation and identification of aptamers from defibrotide that act as thrombin antagonists in vitro. Biochem Biophys Res Commun. 1994;200(2):933–937.
  • Pescador R, Capuzzi L, Mantovani M, et al. Defibrotide: properties and clinical use of an old/new drug. Vascul Pharmacol. 2013;59(1–2):1–10.
  • Corbacioglu S, Greil J, Peters C, et al. Defibrotide in the treatment of children with veno-occlusive disease (VOD): a retrospective multicentre study demonstrates therapeutic efficacy upon early intervention. Bone Marrow Transplant. 2004;33(2):189–195.
  • Palomo M, Mir E, Rovira M, et al. What is going on between defibrotide and endothelial cells? Snapshots reveal the hot spots of their romance. Blood. 2016;127(13):1719–1727.
  • Palomo M, Diaz-Ricart M, Rovira M, et al. Defibrotide prevents the activation of macrovascular and microvascular endothelia caused by soluble factors released to blood by autologous hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2011;17(4):497–506.
  • Eissner G, Multhoff G, Gerbitz A, et al. Fludarabine induces apoptosis, activation, and allogenicity in human endothelial and epithelial cells: protective effect of defibrotide. Blood. 2002;100(1):334–340.
  • Schroder H. Defibrotide protects endothelial cells, but not L929 tumour cells, from tumour necrosis factor-alpha-mediated cytotoxicity. J Pharm Pharmacol. 1995;47(3):250–252.
  • Scalia R, Kochilas L, Campbell B, et al. Effects of defibrotide on leukocyte-endothelial cell interaction in the rat mesenteric vascular bed: role of P-selectin. Methods Find Exp Clin Pharmacol. 1996;18(10):669–676.
  • Richardson PG, Soiffer RJ, Antin JH, et al. Defibrotide for the treatment of severe hepatic veno-occlusive disease and multiorgan failure after stem cell transplantation: a multicenter, randomized, dose-finding trial. Biol Blood Marrow Transplant. 2010;16(7):1005–1017.
  • Falanga A, Vignoli A, Marchetti M, et al. Defibrotide reduces procoagulant activity and increases fibrinolytic properties of endothelial cells. Leukemia. 2003;17(8):1636–1642.
  • Benimetskaya L, Wu S, Voskresenskiy AM, et al. Angiogenesis alteration by defibrotide: implications for its mechanism of action in severe hepatic veno-occlusive disease. Blood. 2008;112(10):4343–4352.
  • Richardson PG, Corbacioglu S, Ho VT, et al. Drug safety evaluation of defibrotide. Expert Opin Drug Saf. 2013;12(1):123–136.
  • Richardson PG, Elias AD, Krishnan A, et al. Treatment of severe veno-occlusive disease with defibrotide: compassionate use results in response without significant toxicity in a high-risk population. Blood. 1998;92(3):737–744.
  • Bonomini V, Frasca GM, Raimondi C, et al. Effect of a new antithrombotic agent (defibrotide) in acute renal failure due to thrombotic microangiopathy. Nephron. 1985;40(2):195–200.
  • Bonomini V, Vangelista A, Frasca G. A new antithrombotic agent in the treatment of acute renal failure due to hemolytic-uremic syndrome and thrombotic thrombocytopenic purpura. Nephron. 1984;37(2):144.
  • Ulutin ON. Antithrombotic effect and clinical potential of defibrotide. Semin Thromb Hemost. 1993;19(Suppl 1):186–191.
  • Tocchetti P, Tudone E, Marier J-F, et al. Pharmacokinetic profile of defibrotide in patients with renal impairment. Drug Des Devel Ther. 2016;10:2631–2641.
  • Chopra R, Eaton JD, Grassi A, et al. Defibrotide for the treatment of hepatic veno-occlusive disease: results of the European compassionate-use study. Br J Haematol. 2000;111(4):1122–1129.
  • Richardson PG, Murakami C, Jin Z, et al. Multi-institutional use of defibrotide in 88 patients after stem cell transplantation with severe veno-occlusive disease and multisystem organ failure: response without significant toxicity in a high-risk population and factors predictive of outcome. Blood. 2002;100(13):4337–4343.
  • Richardson PG, Grupp SA, Pagliuca A, et al. Defibrotide for the treatment of hepatic veno-occlusive disease/sinusoidal obstruction syndrome with multiorgan failure. Int J Hematol Oncol. 2017;6(3):75–93.
  • Corbacioglu S, Carreras E, Mohty M, et al. Defibrotide for the treatment of hepatic veno-occlusive disease: final results from the international compassionate-use program. Biol Blood Marrow Transplant. 2016;22(10):1874–1882.
  • Richardson PG, Riches ML, Kernan NA, et al. Phase 3 trial of defibrotide for the treatment of severe veno-occlusive disease and multi-organ failure. Blood. 2016;127(13):1656–1665.
  • Richardson PG, Smith AR, Triplett BM, et al. Earlier defibrotide initiation post-diagnosis of veno-occlusive disease/sinusoidal obstruction syndrome improves day +100 survival following hematopoietic stem cell transplantation. Br J Haematol. 2017;178(1):112–118.
  • Strouse C, Richardson P, Prentice G, et al. Defibrotide for treatment of severe veno-occlusive disease in pediatrics and adults: an exploratory analysis using data from the Center for International Blood and Marrow Transplant Research. Biol Blood Marrow Transplant. 2016;22(7):1306–1312.
  • Corbacioglu S, Grupp SA, Pagliuca A, et al. Protocol for a phase 3, randomized, adaptive study comparing the efficacy and safety of defibrotide vs best supportive care to prevent hepatic veno-occlusive disease/sinusoidal obstruction syndrome in adult and pediatric patients undergoing hematopoietic stem cell transplant. In: Poster presented at: 2016 American Society of Hematology Meeting on Hematologic Malignancies; 2016 Sep 16–17; Chicago (IL). Poster 8.
  • Palmer KJ, Goa KL. Defibrotide. A review of its pharmacodynamic and pharmacokinetic properties, and therapeutic use in vascular disorders. Drugs. 1993;45(2):259–294.
  • European Medicines Agency. Assessment report – defitelio. In: Committee for medical products for human use. London, UK: CHMP; 2013.
  • US Food and Drug Administration. FDA approves first treatment for rare disease in patients who receive stem cell transplant from blood or bone marrow. Silver Spring, MD: FDA; 2016. FDA news release.
  • Tekgunduz E, Kaya AH, Bozdag SC, et al. Does defibrotide prophylaxis decrease the risk of acute graft versus host disease following allogeneic hematopoietic cell transplantation? Transfus Apher Sci. 2016;54(1):30–34.
  • An Open-Label Study of Defibrotide for the Prevention of Acute Graft-versus-Host-Disease (AGVHD) [Internet]. Bethesda (MD): US National Library of Medicine; [updated 2017 Dec 1; cited 2017 Dec 22]. Available from: https://clinicaltrials.gov/show/NCT03339297

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.