Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Feb 8.
Published in final edited form as: Curr Opin Hematol. 2021 Nov 1;28(6):408–416. doi: 10.1097/MOH.0000000000000687

We have enclosed our article titled “Emerging Therapeutic and Preventative Approaches to Transplant-Associated Thrombotic Microangiopathy”.

Michelle Schoettler 1, Satheesh Chonat 2, Kirsten Williams 1, Leslie Lehmann 3
PMCID: PMC9908033  NIHMSID: NIHMS1737404  PMID: 34534983

Abstract

Purpose of Review:

Transplant associated thrombotic microangiopathy (TA-TMA) is a complication which can occur in both allogeneic and autologous hematopoietic cellular therapy (HCT) recipients and is associated with significant morbidity and mortality. While TA-TMA is a complex disease, there is emerging evidence that complement activation and endothelial dysfunction play a key role in the pathophysiology of the disease. The use of eculizumab has improved survival in patients with high risk and severe disease, but mortality rates in treated patients still exceed 30%, highlighting the need for novel approaches.

Recent Findings:

There are multiple ongoing and planned clinical trials investigating novel complement agents in TA-TMA and other TMAs. Drugs vary by targets of the complement system, mechanism, and form of administration. Clinical trial designs include single arm studies that span across multiple age groups including children, and double blind, randomized, placebo-controlled studies. These studies will provide robust data to inform the care of patients with TA-TMA in the future. In addition to multiple promising therapeutic agents, preventing TA-TMA is an emerging strategy. Agents known to protect the endothelium from damage and augment endothelial function by promoting anti-inflammatory and anti-thrombotic effects may have a role in preventing TA-TMA or ameliorating the severity, though additional studies are needed.

Summary:

Novel therapeutic agents for TA-TMA inhibition of the complement system are under investigation and prophylactic strategies of endothelial protection are emerging. Further understanding of the pathophysiology of the disease may identify additional therapeutic targets. Multi-institutional, collaborative clinical trials are needed to determine the safety and efficacy of these agents going forward.

Keywords: Transplant associated thrombotic microangiopathy (TA-TMA), hematopoietic cellular therapy (HCT), complement inhibition, endothelial dysfunction

Background

Transplant associated thrombotic microangiopathy (TA-TMA) is an increasingly recognized complication of allogeneic and autologous hematopoietic cellular therapy (HCT) associated with significant morbidity and mortality1*,2*,3. TA-TMA shares the defining features of all TMAs: microangiopathic hemolytic anemia, thrombocytopenia, and organ dysfunction. Clinical manifestations of TA-TMA include hypertension, serositis, pulmonary hypertension, diffuse alveolar hemorrhage (DAH), renal insufficiency, posterior reversible encephalopathy syndrome (PRES) and gastrointestinal bleeding3,4*. About 50% or more patients with TA-TMA develop a severe phenotype with multiorgan dysfunction that can progress to organ failure and death5. TA-TMA is primarily a clinical diagnosis as renal biopsies are often not obtained in suspected TA-TMA and there is no consensus on histologic criteria in other organs. While multiple diagnostic criteria have been proposed, the consensus definition requires that ≥4/7 features are met at 2 different time points: hemolytic anemia (or increased need for red blood cell transfusions), de novo thrombocytopenia (or increased need for platelet transfusions), elevated lactate dehydrogenase (LDH), schistocytes on the peripheral blood smear, proteinuria of ≥30 mg/dL in random urine, hypertension defined as ≥99th percentile for age or greater than 140/90 in adults, and elevated soluble membrane attack complex (sC5b-9, ≥ 244 ng/mL)6,7. A high degree of clinical suspicion is required to detect TA-TMA before the development of multiorgan dysfunction as these laboratory and clinical findings may be caused by other common HCT complications.

Given inherent limitations of the clinical diagnostic criteria and different screening practices, the incidence of TA-TMA varies. TA-TMA is thought to complicate 10–35% of allogeneic HCTs2 and ~25% of pediatric autologous HCTs in children with neuroblastoma8,9. Risk factors for TA-TMA can be grouped into patient characteristics, transplant characteristics, and complications post HCT. Patient characteristic risks for TA-TMA include: underlying diagnosis of HLH10, aplastic anemia11, female sex12, non-white race, and genetic variants in complement proteins13. Transplant characteristics include a second allogeneic HCT14, myeloablative conditioning regimen15, and HLA disparity16 and post-transplant complications of bacterial, viral and/or fungal infections2, acute graft versus host disease (aGVHD)14, and the use of calcineurin inhibitors (CNI)17, particularly when combined with sirolimus18. This has led to the “three hit hypothesis” of TA-TMA genesis: hit 1) a predisposing endothelial injury and/or an underlying predisposition to complement activation, hit 2) receipt of chemotherapy which further damages the endothelium, and hit 3) additional insults like aGVHD or infection4*.

TA-TMA confers an increased non-relapse mortality rate and decreased overall survival compared to patients without TA-TMA7,19,20. In a prospective cohort, elevated sC5b-9 and proteinuria at the time of TA-TMA diagnosis was prognostic and associated with a 1-year OS rate of 16.7%, compared to 1-year survival rate of 100% in children with TA-TMA who did not have these features7. Thus, these markers are used most commonly for prognosis and treatment decisions. The “TMA index” (LDH IU/L divided by platelet count) is also prognostic, with patients scoring >100 having very poor outcomes21.

The pathophysiology of TA-TMA is not fully understood. It is thought to start with endothelial damage, leading to endothelial activation and a pro-coagulant, pro-inflammatory state with activation of complement and generation of microthrombi, leading to ongoing endothelial cell activation (Figure 2). Pathways in endothelial cell activation are largely derived from work in atherosclerosis, but there are some biomarker studies in TA-TMA patients that support similar processes22. The role of complement in the pathophysiology of the disease is suggested by an increased risk of TA-TMA in patients with complement genetic mutations13, perturbation in complement regulatory function, elevated levels of MASP-223**, significant increases in C3b and sC5b-924, and the therapeutic effect of eculizumab, a monoclonal antibody to C5. In a study of pediatric patients with high-risk TA-TMA (elevated sC5b-9 and proteinuria), the 1-year overall survival of patients treated with eculizumab was 66% compared to 16.7% in historical (untreated) controls25**. Much of the literature on eculizumab use in TA-TMA is in pediatric cohorts. While responses to eculizumab have been reported in adults with TA-TMA, survival remains poor26,27. Regardless of treatment, survivors of TA-TMA, have increased morbidity due to chronic kidney disease20, persistent pulmonary hypertension28, and neurologic sequelae29.

Figure 2: Proposed pathophysiology of TA-TMA and potential therapeutic targets.

Figure 2:

Proposed Pathophysiology of TA-TMA: Endothelial cells are damaged by chemotherapy given as part of the BMT, medications, and complications of BMT. Damaged ECs release ANG-2, leading to vascular destabilization, lead to decreased nitric oxide production (NO) and releases IL-8. Local recruitment of antigen presenting cells (APCs) and lymphocytes occurs due to the expression of increased adhesion molecules by activated APCs. APCs also express TNF-alpha, INF-gamma, IL-6, IL-8 and IL-12, further stimulating activation of T-cells and monocytes/macrophages. This stimulates neutrophils to release neutrophil extracellular traps (NETs). Decreased levels of NO leads to loss of suppression of complement and NETs activate complement. Activation of C3 leads to the formation of C3b, which binds to the endothelial surface leading to formation of the membrane attack complex (MAC) on the endothelial cells, causing additional endothelial cell damage. Tissue factor (TF), which is expressed on the cell surface binds factor VIIIA and von Willebrand factor (vWF) promoting activated platelets and subsequent thrombus formation. Microthrombi then lead to organ ischemia and end organ damage (e.g. renal failure). Potential prophylactic and therapeutic targets and agents are indicated. Figure was created using biorender.com.

Current Management Paradigm

The current management paradigm for TA-TMA first emphasizes preventing (when possible) and mitigating complications or medications that drive TA-TMA (Figure 1). Treating infections and aGVHD may decrease the severity of TMA by dampening the feedback loop of endothelial damage and inflammation. Removing calcineurin inhibitors (CNIs) may improve TA-TMA, by diminishing the direct toxic effects on the endothelium (studied in vitro)30,31 32. However, this strategy is complicated in patients with concurrent aGVHD. Uncontrolled aGVHD is also a risk factor for the development of TA-TMA, and the period of decreased immunosuppression incurred while switching from a CNI to another agent may increase the severity of GVHD. In a small study, switching GVHD prophylaxis from a CNI to daclizumab resulted in improvement of TA-TMA in some patients; however mortality from aGVHD was unacceptable33.

Figure 1: Current Management Paradigm.

Figure 1:

TA-TMA is currently managed with supportive care of concurrent illnesses and complications, and in patients with high-risk TA-TMA or severe disease, eculizumab. Plasmapheresis and rituximab are generally not effective unless there is an autoantibody present.

Optimizing supportive care measures is recommended for associated complications of TA-TMA including: hypertension, acute kidney injury, pericardial or pleural effusion, PRES, DAH, and gastrointestinal bleeding. If patients have high titers of factor H autoantibodies, treatment with therapeutic plasma exchange (TPE) may be helpful34, and treating with rituximab at the end of TPE can be considered35. If there are no antibodies present, TPE and rituximab have limited efficacy36. Eculizumab is recommended for patients with high risk features defined as elevated sC5b-9 (≥244 ng/mL) and proteinuria (>30 mg/dL)37. Eculizumab may also be considered when patients have organ dysfunction and elevated sC5b-9 or proteinuria, or in those with progressive disease despite maximal supportive care. Eculizumab is dosed frequently early in the course of TA-TMA to achieve suppression of complement, with CH50 used as an indirect measure of complement blockade38. After an early intense treatment period, eculizumab is spaced out over time. If the inciting factor has resolved, i.e., GVHD is quiescent or infection is treated, patients may be able to stop eculizumab without recurrent TA-TMA. More data is needed regarding the length of therapy, patients most likely to benefit, the pharmacokinetics of eculizumab in TA-TMA, and an ongoing phase 2 multi-institutional trial will likely provide important data to this effect (ClinicalTrials.gov: NCT03518203). Although eculizumab has shown success in some patients and generally been safe when prophylaxis against Neisserial bacterial infections are provided, it is expensive and nearly 40% do not respond and die25**. Thus, new approaches are needed. Additional understanding of the pathophysiology, consensus on diagnostic criteria and response, and new diagnostic and prognostic biomarkers are critically important as additional therapeutic approaches emerge.

Emerging Therapeutic Approaches

There are multiple complement inhibitors under investigation that may have therapeutic implications in TA-TMA (Table 1). The agents currently FDA approved for paroxysmal nocturnal hemoglobinuria (PNH) or atypical hemolytic uremic syndrome (aHUS) include: eculizumab, ravulizumab, and pegcetacoplan. Medications with published data or current or planned trials specifically in TA-TMA include:

  • Ravulizumab/Ultomiris® is a long acting humanized monoclonal C5 antibody, currently FDA approved for PNH and aHUS. There are two ongoing clinical trials in TA-TMA, one a phase 3 randomized, double blind, placebo-controlled trial (clinicaltrials.gov NCT04543591) and a phase 3 single arm study in pediatrics (clinicaltrials.gov NCT04557735). This is the first randomized trial in TA-TMA and is a critically important addition to the field of TA-TMA.

  • OMS721/Narsoplimab® is a novel mannan-binding lectin-associated serine protease-2 (MASP-2) pathway inhibitor. A single arm phase 2 study of OMS721 demonstrated improved overall survival in adult patients with TA-TMA compared to a historic cohort and improvement in hematologic parameters23**. The drug received FDA breakthrough approval, and the company has announced commitment to opening a pediatric trial in in the near future.

  • Coversin/ Nomacopan® is a recombinant C5 inhibitor originally derived from the hematophagous tick with ongoing clinical trials in PNH and aHUS. Coversin overcomes variants in C5 that result in eculizumab resistance. In a case report, a child with eculizumab refractory TA-TMA responded to coversin39. The company has announced a plan for clinical trials in both adults and pediatrics.

Table 1:

Complement agents currently with active clinical trials in TMAs or announced upcoming trials in TA-TMA

Agent Target Admin Pharmaceutical Sponsor TMA type Current Active Studies Age
Eculizumab/Soliris ® mAb inhibits C5 IV Alexion Pharmaceuticals TA-TMA* Phase 2 single arm Any age
Ravulizumab/Ultomiris ® mAb inhibits C5 (long acting, same epitope as eculizumab) IV Alexion Pharmaceuticals TA-TMA* Phase 3 Single arm
Phase 3 DB RCT
≥1 month <18 years ≥12 years
Coversin/Nomacopan® Recombinant protein, inhibits C5 & leukotriene B4 SQ Akari Therapeutics TA-TMA# Announced in planning
OMS721/Narsoplimab® Inhibits lectin pathway activation IV Omeros TA-TMA^ Announced in planning
REGN3918 Inhibits C5 SQ Regeneron PNH Phase 3 Single arm Adults
LFG-316/Tesidolumab Inhibits C5 IV Novartis PNH Phase 2 Adults
ABP959 mAb inhibits C5 (biosimilar) IV Amgen PNH Phase 3 DB randomized crossover Study Adults
SKY59/RO7112689/Crovalimab mAb inhibits C5 SQ Hoffmann-La Roche aHUS, PNH Phase 3, single arm ≥12 years
RA101495/Zilucoplan Inhibit C5 SQ Ra Pharmaceuticals PNH Phase 2 Adults
ALN-CC5/Cemdisiran RNAi, silence hepatic C5 production SQ Alnylam PNH Phase 2 Adults
APL2/Pegcetacoplan Pegylated peptide, inhibits C3 SQ Apellis Pharmaceuticals, Inc. PNH Phase 3 DB RCT Adults
LNP023/Iptacopan Small molecule, inhibits factor B, preventing C3 convertase formation oral Novartis Pharmaceuticals aHUS PNH Phase 3 DB RCT Adults
ALXN2040/Danicopan Small molecule, inhibits factor D, preventing C3 convertase formation Oral Alexion Pharmaceuticals PNH Phase 2 Adults
Sutimlimab Inhibits C1 protease activity IV Sanofi CAD Phase 2, Phase 3 DB RCT Adults
*

completed studies in PNH and aHUS

#

completed study in PNH

^

completed study in aHUS

mAb- monoclonal antibody, IV- intravenous, SQ- sub cutaneous, DB RCT- double blind randomized control trial, PNH- paroxysomal nocturnal hemoglobinuria, aHUS- atypical hemolytic uremic syndrome, CAD- cold agglutinin disease

Agents currently investigated for other complement driven diseases, i.e., vasculitis or renal diseases, were not included in this review. Studies that were withdrawn on clinicaltrial.gov were also excluded.

Immune Effector Blockade:

Current understanding of the action of immune effector cells and subsequent inflammatory cytokines at the microvascular level in TA-TMA are derived from data generated primarily from atherosclerosis40 and/or other TMAs41. There is emerging evidence from RNA sequencing data for an INF-gamma and activated complement loop in TA-TMA42*. While elevated serum levels of inflammatory cytokines including TNF-alpha, INF-gamma, and IL-8 are reported in other HCT complications, to our knowledge elevation of different cytokines has not been described in TA-TMA. If these pathways are altered in TA-TMA and contribute to the disease pathogenesis, inhibitors including etanercept (TNF-alpha), infliximab (TNF-alpha), tocilizumab (IL-6), and emapalumab (INF-gamma) may be additional therapeutic options.

Defibrotide:

Defibrotide, an oligonucleotide derived from pig intestines and is currently FDA approved for the treatment of VOD with organ dysfunction. While its mechanism is not fully understood, defibrotide is thought to reduce endothelial activation and enhance fibrinolysis, exerting protective effects on the endothelium. There are retrospective reports using defibrotide to treat TA-TMA with overall survival rates ranging from 57%43 to 77%44. The duration of use of defibrotide varied considerably among the studies. Anecdotally, patients have developed TA-TMA while receiving defibrotide for VOD prophylaxis. Defibrotide is expensive but generally well tolerated and thus further studies evaluating the efficacy of defibrotide for TA-TMA may be warranted.

Recombinant thrombomodulin(rTM):

Thrombomodulin is a transmembrane glycoprotein that exerts antithrombotic, antifibrinolytic, anti-inflammatory, and cytoprotective properties45. Activated neutrophils in endothelial activation cleave thrombomodulin, obviating the vasoprotective properties. rTM has mixed results in multiple clinical trials investigating the impact on sepsis and disseminated intravascular coagulation (DIC)46,47 In small case series or reports, rTM has been used to treat TA-TMA with varying responses48,49. While rTM has biologic features that make it a plausible therapeutic agent, there is currently limited data to support its use in TA-TMA.

Emerging Prophylactic Approaches

Preventing severe illness is easier than treating it, and thus prophylaxis is a keystone approach to multiple HCT complications including GVHD, infections and veno-occlusive disease (VOD). While some risk factors of TA-TMA are not modifiable, like race or donor match, there is evidence that general measures aimed at improving endothelial health are protective against other types of TMAs in animal models50. Thus, there is biologic premise that endothelial protection may be an effective strategy to prevent TA-TMA in retrospective studies and series, in-vitro studies, and animal models (Figure 2).

Defibrotide:

Defibrotide is used for prevention of VOD in high risk HCT recipients5153. In vivo, defibrotide protects endothelial cells against damage from CNI induced apoptosis54 and fludarabine55. There are emerging studies that suggest the use of defibrotide may result in lower rates of acute GVHD56,57 a known risk factor for the development of TA-TMA. Thus, defibrotide could be an effective agent for the prevention of TA-TMA via several mechanisms. The incidence of TA-TMA was not reported in previous clinical trials when defibrotide was administered for VOD prophylaxis. However, there is an ongoing single arm feasibility trial evaluating the use of defibrotide as prophylaxis for TA-TMA in children with high-risk disease (ClinicalTrials.gov NCT03384693). Further studies are warranted to investigate the role of defibrotide prophylaxis in TA-TMA and the effect if given only in the immediate post HCT period.

Vitamin D:

There is emerging data that Vitamin D plays a role in endothelial health by augmenting nitric oxide production and reducing IL-6 and vascular and intravascular cell adhesion molecule expression (VCAM and ICAM)58. Vitamin D deficiency is linked to endothelial dysfunction as measured by flow mediated dilation59 and linked to cardiovascular disease and atherosclerosis, also endothelial diseases. In-vitro vitamin D protects endothelial cells from radiation induced apoptosis60. Some though not all prospective studies have shown vitamin D deficiency is associated with increased risk of death after HCT61,62. To our knowledge, there are no published data to evaluate vitamin D deficiency prior to HCT as a risk factor for subsequent TA-TMA, though a phase 4 clinical trial is ongoing to test the safety and efficacy of a single high oral dose of vitamin D pre-HCT in children (Clinicaltrial.gov NCT03176849); secondary endpoints will capture the incidence of transplant complications. Given low cost, minimal side effects, and benefit to bone health, checking vitamin D levels prior to HCT and repleting deficiencies should likely be incorporated into practice, though the effect on TA-TMA risk is unknown.

Statins:

While the indication for statins is hypercholesteremia, they are also thought to improve endothelial cell function by increasing the bioavailability of nitric oxide, promoting reendothelialization, reducing oxidative stress, and inhibiting inflammatory responses63. In an study in adult HCT, with retrospective diagnosis of TA-TMA, a multivariable analysis showed the use of pravastatin and ursodiol was associated with a decreased incidence of TA-TMA compared to an untreated historic cohort (HR 0.23, 95% CI 0.11–0.49)64. Statins are relatively well tolerated with minimal side effects, and regularly used in the adult setting. Statins have limited dose formulations available, which could make their use in the pediatric setting challenging. Further, statins have multiple medication interactions which may complicate their use in the immediate peri-HCT period. Additional studies on the efficacy and safety of this approach are needed.

Eicosapentaenoic acid (EPA):

EPA is an omega-3 fatty acid thought to inhibit thrombosis, augment endothelial vasodilation via nitric oxide and suppress the production of inflammatory cytokines. In a small study, patients who were given EPA from day −21 to day +180 had a lower incidence of TA-TMA and improved survival compared to patients who did not receive EPA65.

N-acetyl cysteine (NAC):

NAC is an antioxidant that removes reactive oxygen species by increasing glutathione biosynthesis. In a mouse model of shiga-toxin-induced hemolytic uremic syndrome, the administration of NAC was partially protective against the development of renal failure and death66. In a retrospective study, a combination of EPA and NAC was administered to patients classified as high risk for developing TA-TMA (high risk if 3 of the following criteria are met: age>10 years, race ethnicity non-white, ABO blood group minor incompatibility, or haploidentical donor). Compared to historic high-risk controls, the incidence of TA-TMA among the EPA/NAC treated group decreased from 28.2% to 4.5%11*. A phase 3, randomized, double blind placebo-controlled trial investigating NAC for prophylaxis of TA-TMA is ongoing (clinicaltrial.gov NCT03252925).

Complement blockade:

Agents that inhibit complement proteins are the leading area of growth in emerging therapeutic agents, but this strategy as a prophylactic approach has not been trialed. There is some evidence that elevation of sC5b-9 on day 28 compared to baseline levels prior to HCT are associated with the later development of TA-TMA67. However, the effect (if any) of blocking complement pre-emptively is not known. This approach is expensive and currently lacks data. Additional investigation is needed to determine if activated complement early in the course of HCT is associated with later development of TA-TMA.

Conclusions

TA-TMA is associated with significant morbidity and mortality, and new approaches are needed. There are many promising novel complement blockade agents under investigation, and emerging approaches to prophylaxis. Further understanding of the pathophysiology and risk factors for the disease will inform additional prophylactic and therapeutic options. This field would benefit from specific consensus on diagnostic criteria and markers of prognosis to best define the population who would most benefit from novel therapies and pre-emptive approaches. Multi-institutional, collaborative clinical trials are then needed to develop these criteria, understand incidence and risk factors, and to critically evaluate the efficacy of therapies and side effects.

Key Points:

  • Transplant associated thrombotic microangiopathy (TA-TMA) is a common, potentially severe complication of hematopoietic cellular therapy (HCT) thought to be driven by activated complement and endothelial dysfunction.

  • Current standard of care in patients with high risk or severe disease includes supportive care and inhibition of the complement system with eculizumab, a monoclonal antibody to C5. While survival has improved with this approach, mortality rates still exceed 30%.

  • Multiple clinical trials are announced or currently enrolling children and adults investigating novel complement inhibition agents in TA-TMA and other TMAs with complement activation.

  • In addition to novel therapeutic agents, prophylactic approaches are emerging and merit further study.

Footnotes

This manuscript has not been published elsewhere and is original. All authors substantially contributed. In line with the aim of Current Opinion articles, we provide an expert review in this subject and have highlighted key recent articles in this field. MS, KW, and LL have no conflicts of interest to disclose. SC is a consultant to Alexion, Agios, Novartis, and Takeda. All tables and figures are original.

References

  • 1.Dandoy CE, Rotz S, Alonso PB, et al. A pragmatic multi-institutional approach to understanding transplant-associated thrombotic microangiopathy after stem cell transplant. Blood Adv. 2020;5(1):1–11. doi: 10.1182/bloodadvances.2020003455 [DOI] [PMC free article] [PubMed] [Google Scholar]; * This is the first multiinstitutional report of the incidence of TA-TMA.
  • 2.Schoettler M, Lehmann LE, Margossian S, et al. Risk factors for transplant-associated thrombotic microangiopathy and mortality in a pediatric cohort. Blood Adv. 2020;4(11):2536–2547. doi: 10.1182/bloodadvances.2019001242 [DOI] [PMC free article] [PubMed] [Google Scholar]; * This paper highlights that TA-TMA likely underappreciated.
  • 3.Laskin BL, Goebel J, Davies SM, Jodele S. Small vessels, big trouble in the kidneys and beyond: hematopoietic stem cell transplantation–associated thrombotic microangiopathy. Blood. 2011;118(6):1452 LP–1462. doi: 10.1182/blood-2011-02-321315 [DOI] [PubMed] [Google Scholar]
  • 4.Dvorak CC, Higham C, Shimano KA. Transplant-Associated Thrombotic Microangiopathy in Pediatric Hematopoietic Cell Transplant Recipients: A Practical Approach to Diagnosis and Management. Front Pediatr. 2019;7:133. https://www.frontiersin.org/article/10.3389/fped.2019.00133. [DOI] [PMC free article] [PubMed] [Google Scholar]; *This paper proposes the “three hit hypothesis”, which has implications in novel therapeutic and prophylactic approaches in TA-TMA
  • 5.Jodele S, Davies SM, Lane A, et al. Diagnostic and risk criteria for HSCT-associated thrombotic microangiopathy: a study in children and young adults. Blood. 2014;124(4):645 LP–653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Uderzo C, Jodele S, El Missiry M, Cicerci F, Busca A, Bacigalupa ACS. Transplant-Associated Thrombotic Microangiopathy (TA-TMA) and Consensus Based Diagnostic and Therapeutic Recommendations: Which TA-TMA Patients to Treat and When? J Bone Marrow Res. 2014;02(03). doi: 10.4172/2329-8820.1000152 [DOI] [Google Scholar]
  • 7.Jodele S, Davies SM, Lane A, et al. Diagnostic and risk criteria for HSCT-associated thrombotic microangiopathy: a study in children and young adults. Blood. 2014;124(4):645 LP–653. http://www.bloodjournal.org/content/124/4/645.abstract. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Jodele S, Dandoy CE, Myers K, et al. High-dose Carboplatin/Etoposide/Melphalan increases risk of thrombotic microangiopathy and organ injury after autologous stem cell transplantation in patients with neuroblastoma. Bone Marrow Transplant. 2018. doi: 10.1038/s41409-018-0159-8 [DOI] [PubMed] [Google Scholar]
  • 9.Laskin BL, Goebel J, Davies SM, et al. Early clinical indicators of transplant-associated thrombotic microangiopathy in pediatric neuroblastoma patients undergoing auto-SCT. Bone Marrow Transplant. 2011;46(5):682–689. doi: 10.1038/bmt.2010.182 [DOI] [PubMed] [Google Scholar]
  • 10.Gloude NJ, Dandoy CE, Davies SM, et al. Thinking Beyond HLH: Clinical Features of Patients with Concurrent Presentation of Hemophagocytic Lymphohistiocytosis and Thrombotic Microangiopathy. J Clin Immunol. 2020;40(5):699–707. doi: 10.1007/s10875-020-00789-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Higham CS, Collins G, Shimano KA, et al. Transplant-associated thrombotic microangiopathy in pediatric patients: pre-HSCT risk stratification and prophylaxis. Blood Adv. 2021;5(8):2106–2114. doi: 10.1182/bloodadvances.2020003988 [DOI] [PMC free article] [PubMed] [Google Scholar]; * This is one of the few papers reporting a prophylactic approach to TA-TMA and has some promising data.
  • 12.Epperla N, Li A, Logan B, et al. Incidence, Risk Factors for and Outcomes of Transplant-Associated Thrombotic Microangiopathy. Br J Haematol. 2020;189(6):1171–1181. doi: 10.1111/bjh.16457 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Jodele S, Zhang K, Zou F, et al. The genetic fingerprint of susceptibility for transplant associated thrombotic microangiopathy. Blood. 2015;127(8):989–997. doi: 10.1182/blood-2015-08-663435 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Elfeky R, Lucchini G, Lum S-H, et al. New insights into risk factors for transplant-associated thrombotic microangiopathy in pediatric HSCT. Blood Adv. 2020;4(11):2418–2429. doi: 10.1182/bloodadvances.2019001315 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Willems E, Baron F, Seidel L, Frère P, Fillet G, Beguin Y. Comparison of thrombotic microangiopathy after allogeneic hematopoietic cell transplantation with high-dose or nonmyeloablative conditioning. Bone Marrow Transplant. 2010;45(4):689–693. doi: 10.1038/bmt.2009.230 [DOI] [PubMed] [Google Scholar]
  • 16.Matsui H, Arai Y, Imoto H, et al. Risk factors and appropriate therapeutic strategies for thrombotic microangiopathy after allogeneic HSCT. Blood Adv. 2020;4(13):3169–3179. doi: 10.1182/bloodadvances.2020002007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Labrador J, López-Corral L, López-Godino O, et al. Risk factors for thrombotic microangiopathy in allogeneic hematopoietic stem cell recipients receiving GVHD prophylaxis with tacrolimus plus MTX or sirolimus. Bone Marrow Transplant. 2014;49(5):684–690. doi: 10.1038/bmt.2014.17 [DOI] [PubMed] [Google Scholar]
  • 18.Rodriguez R, Nakamura R, Palmer JM, et al. A phase II pilot study of tacrolimus/sirolimus GVHD prophylaxis for sibling donor hematopoietic stem cell transplantation using 3 conditioning regimens. Blood. 2010;115(5):1098–1105. doi: 10.1182/blood-2009-03-207563 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Cho B-S, Min C-K, Eom K-S, et al. Clinical impact of thrombotic microangiopathy on the outcome of patients with acute graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant. 2008;41(9):813–820. doi: 10.1038/sj.bmt.1705976 [DOI] [PubMed] [Google Scholar]
  • 20.Postalcioglu M, Kim HT, Obut F, et al. Impact of Thrombotic Microangiopathy on Renal Outcomes and Survival after Hematopoietic Stem Cell Transplantation. Biol Blood Marrow Transplant. 2018;24(11):2344–2353. doi: 10.1016/j.bbmt.2018.05.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Uderzo C, Bonanomi S, Busca A, et al. Risk factors and severe outcome in thrombotic microangiopathy after allogeneic hematopoietic stem cell transplantation. Transplantation. 2006;82(5):638–644. doi: 10.1097/01.tp.0000230373.82376.46 [DOI] [PubMed] [Google Scholar]
  • 22.Anthony MT, Zeigler ZR, Lister J, et al. Plasminogen activator inhibitor (PAI-1) antigen levels in primary TTP and secondary TTP post-bone marrow transplantation. Am J Hematol. 1998;59(1):9—14. doi: [DOI] [PubMed] [Google Scholar]
  • 23.Elhadad S, Chapin J, Copertino D, Van Besien K, Ahamed J, Laurence J. MASP2 levels are elevated in thrombotic microangiopathies: association with microvascular endothelial cell injury and suppression by anti-MASP2 antibody narsoplimab. Clin Exp Immunol. 2021;203(1):96–104. doi: 10.1111/cei.13497 [DOI] [PMC free article] [PubMed] [Google Scholar]; **This is the first clinical trial data from a novel complement inhibitor for TA-TMA. These data are very promising, and are the premise for the potential therapeutic benefic of other complement inhibitor agents
  • 24.Qi J, Wang J, Chen J, et al. Plasma levels of complement activation fragments C3b and sC5b-9 significantly increased in patients with thrombotic microangiopathy after allogeneic stem cell transplantation. Ann Hematol. 2017;96(11):1849–1855. doi: 10.1007/s00277-017-3092-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Jodele S, Dandoy CE, Lane A, et al. Complement blockade for TA-TMA: lessons learned from a large pediatric cohort treated with eculizumab. Blood. 2020;135(13):1049–1057. doi: 10.1182/blood.2019004218 [DOI] [PMC free article] [PubMed] [Google Scholar]; **This is the largest study of the use of eculizumab in TA-TMA.
  • 26.Rudoni J, Jan A, Hosing C, Aung F, Yeh J. Eculizumab for transplant-associated thrombotic microangiopathy in adult allogeneic stem cell transplant recipients. Eur J Haematol. 2018;0(0). doi: 10.1111/ejh.13127 [DOI] [PubMed] [Google Scholar]
  • 27.Bohl SR, Harsdorf S, Schoensteiner S, et al. Eculizumab Therapy of Adult TA-TMA: A High Response Rate Is Associated with a High Infection-Related Mortality. Blood. 2016;128(22):2255 LP–2255. http://www.bloodjournal.org/content/128/22/2255.abstract. [Google Scholar]
  • 28.Dandoy CE, Hirsch R, Chima R, Davies SM, Jodele S. Pulmonary hypertension after hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2013;19(11):1546–1556. doi: 10.1016/j.bbmt.2013.07.017 [DOI] [PubMed] [Google Scholar]
  • 29.Schoettler M, Duncan C, Lehmann L. Severe, persistent neurotoxicity after transplant-associated thrombotic microangiopathy in a pediatric patient despite treatment with eculizumab. Pediatr Transplant. 2019;23(3). doi: 10.1111/petr.13381 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Carmona A, Díaz-Ricart M, Palomo M, et al. Distinct Deleterious Effects of Cyclosporine and Tacrolimus and Combined Tacrolimus–Sirolimus on Endothelial Cells: Protective Effect of Defibrotide. Biol Blood Marrow Transplant. 2013;19(10):1439–1445. doi: 10.1016/j.bbmt.2013.07.001 [DOI] [PubMed] [Google Scholar]
  • 31.Li A, Wu Q, Davis C, et al. Transplant-Associated Thrombotic Microangiopathy Is a Multifactorial Disease Unresponsive to Immunosuppressant Withdrawal. Biol Blood Marrow Transplant. 2019;25(3):570–576. doi: 10.1016/j.bbmt.2018.10.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Kanunnikov MM, Rakhmanova ZZ, Levkovsky NV, et al. Conversion from calcineurin inhibitors to sirolimus in transplant-associated thrombotic microangiopathy. Clin Transplant. 2021;35(2):e14180. doi: 10.1111/ctr.14180 [DOI] [PubMed] [Google Scholar]
  • 33.Wolff D, Wilhelm S, Hahn J, et al. Replacement of calcineurin inhibitors with daclizumab in patients with transplantation-associated microangiopathy or renal insufficiency associated with graft-versus-host disease. Bone Marrow Transplant. 2006;38(6):445–451. doi: 10.1038/sj.bmt.1705454 [DOI] [PubMed] [Google Scholar]
  • 34.Khandelwal P, Gupta A, Sinha A, et al. Effect of plasma exchange and immunosuppressive medications on antibody titers and outcome in anti-complement factor H antibody-associated hemolytic uremic syndrome. Pediatr Nephrol. 2015;30(3):451–457. doi: 10.1007/s00467-014-2948-7 [DOI] [PubMed] [Google Scholar]
  • 35.Dragon-Durey M-A, Loirat C, Cloarec S, et al. Anti–Factor H Autoantibodies Associated with Atypical Hemolytic Uremic Syndrome. J Am Soc Nephrol. 2005;16(2):555 LP–563. doi: 10.1681/ASN.2004050380 [DOI] [PubMed] [Google Scholar]
  • 36.Sartain S, Shubert S, Wu MF, et al. Therapeutic Plasma Exchange does not Improve Renal Function in Hematopoietic Stem Cell Transplantation–Associated Thrombotic Microangiopathy: An Institutional Experience. Biol Blood Marrow Transplant. 2018;000:1–6. doi: 10.1016/j.bbmt.2018.08.016 [DOI] [PubMed] [Google Scholar]
  • 37.Sonata Jodele CUC. Transplant-Associated Thrombotic Microangiopathy (TA-TMA) and Consensus Based Diagnostic and Therapeutic Recommendations: Which TA-TMA Patients to Treat and When? J Bone Marrow Res. 2014;02(03). doi: 10.4172/2329-8820.1000152 [DOI] [Google Scholar]
  • 38.Jodele S, Fukuda T, Mizuno K, et al. Variable eculizumab clearance requires pharmacodynamic monitoring to optimize therapy for thrombotic microangiopathy after hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2016;22(2):307–315. doi: 10.1016/j.bbmt.2015.10.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Goodship THJ, Pinto F, Weston-Davies WH, et al. Use of the complement inhibitor Coversin to treat HSCT-associated TMA. Blood Adv. 2017;1(16):1254–1258. doi: 10.1182/bloodadvances.2016002832 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Jean D, Peter G. Role of Endothelial Dysfunction in Atherosclerosis. Circulation. 2004;109(23_suppl_1):III-27–III-32. doi: 10.1161/01.CIR.0000131515.03336.f8 [DOI] [PubMed] [Google Scholar]
  • 41.Westwood JP, Langley K, Heelas E, Machin SJ, Scully M. Complement and cytokine response in acute Thrombotic Thrombocytopenic Purpura. Br J Haematol. 2014;164(6):858–866. doi: 10.1111/bjh.12707 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Jodele S, Medvedovic M, Luebbering N, et al. Interferon-complement loop in transplant-associated thrombotic microangiopathy. Blood Adv. 2020;4(6):1166–1177. doi: 10.1182/bloodadvances.2020001515 [DOI] [PMC free article] [PubMed] [Google Scholar]; *This provides premise and biologic plausability for INF-gamma blockade as a novel therapeutic option in TA-TMA
  • 43.Corti P, Uderzo C, Tagliabue A, et al. Defibrotide as a promising treatment for thrombotic thrombocytopenic purpura in patients undergoing bone marrow transplantation. Bone Marrow Transplant. 2002;29(6):542–543. doi: 10.1038/sj.bmt.1703414 [DOI] [PubMed] [Google Scholar]
  • 44.Yeates L, Slatter MA, Bonanomi S, et al. Use of defibrotide to treat transplant-associated thrombotic microangiopathy: A retrospective study of the Paediatric Diseases and Inborn Errors Working Parties of the European Society of Blood and Marrow Transplantation. Bone Marrow Transplant. 2017;52(5):762–764. doi: 10.1038/bmt.2016.351 [DOI] [PubMed] [Google Scholar]
  • 45.Loghmani H, Conway EM. Exploring traditional and nontraditional roles for thrombomodulin. Blood. 2018;132(2):148–158. doi: 10.1182/blood-2017-12-768994 [DOI] [PubMed] [Google Scholar]
  • 46.Vincent J-L, Francois B, Zabolotskikh I, et al. Effect of a Recombinant Human Soluble Thrombomodulin on Mortality in Patients With Sepsis-Associated Coagulopathy: The SCARLET Randomized Clinical Trial. JAMA. 2019;321(20):1993–2002. doi: 10.1001/jama.2019.5358 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Mori S, Ai T, Sera T, Ochiai K, Otomo Y. Human Soluble Recombinant Thrombomodulin, ART-123, Resolved Early Phase Coagulopathies, but Did Not Significantly Alter the 28 Day Outcome in the Treatment of DIC Associated with Infectious Systemic Inflammatory Response Syndromes. J Clin Med. 2019;8(10):1553. doi: 10.3390/jcm8101553 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Fujiwara H, Maeda Y, Sando Y, et al. Treatment of thrombotic microangiopathy after hematopoietic stem cell transplantation with recombinant human soluble thrombomodulin. Transfusion. 2016;56(4):886–892. doi: 10.1111/trf.13437 [DOI] [PubMed] [Google Scholar]
  • 49.Arai Y, Yamashita K, Mizugishi K, et al. Serum Neutrophil Extracellular Trap Levels Predict Thrombotic Microangiopathy after Allogeneic Stem Cell Transplantation. Biol Blood Marrow Transplant. 2013;19(12):1683–1689. doi: 10.1016/J.BBMT.2013.09.005 [DOI] [PubMed] [Google Scholar]
  • 50.Goldberg RJ, Nakagawa T, Johnson RJ, Thurman JM. The Role of Endothelial Cell Injury in Thrombotic Microangiopathy. Am J Kidney Dis. 2010;56(6):1168–1174. doi: 10.1053/j.ajkd.2010.06.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.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. doi: 10.1016/j.bbmt.2010.02.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.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. doi: 10.1038/sj.bmt.1704329 [DOI] [PubMed] [Google Scholar]
  • 53.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. 2012;379(9823):1301–1309. doi: 10.1016/S0140-6736(11)61938-7 [DOI] [PubMed] [Google Scholar]
  • 54.Wang X, Pan B, Hashimoto Y, et al. Defibrotide Stimulates Angiogenesis and Protects Endothelial Cells from Calcineurin Inhibitor-Induced Apoptosis via Upregulation of AKT/Bcl-xL. Thromb Haemost. 2018;118(02):161–174. [DOI] [PubMed] [Google Scholar]
  • 55.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. doi: 10.1182/blood.V100.1.334 [DOI] [PubMed] [Google Scholar]
  • 56.Tekgündüz E, Kaya AH, Bozdağ 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. doi: 10.1016/j.transci.2016.01.009 [DOI] [PubMed] [Google Scholar]
  • 57.García-Bernal D, Palomo M, Martínez CM, et al. Defibrotide inhibits donor leucocyte-endothelial interactions and protects against acute graft-versus-host disease. J Cell Mol Med. 2020;24(14):8031–8044. doi: 10.1111/jcmm.15434 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Dalan R, Liew H, Tan WKA, Chew DEK, Leow MK-S. Vitamin D and the endothelium: basic, translational and clinical research updates. IJC Metab Endocr. 2014;4:4–17. doi: 10.1016/j.ijcme.2014.06.003 [DOI] [Google Scholar]
  • 59.Mazidi M, Karimi E, Rezaie P, Vatanparast H. The impact of vitamin D supplement intake on vascular endothelial function; a systematic review and meta-analysis of randomized controlled trials. Food Nutr Res. 2017;61(1):1273574. doi: 10.1080/16546628.2016.1273574 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Marampon F, Gravina GL, Festuccia C, et al. Vitamin D protects endothelial cells from irradiation-induced senescence and apoptosis by modulating MAPK/SirT1 axis. J Endocrinol Invest. 2016;39(4):411–422. doi: 10.1007/s40618-015-0381-9 [DOI] [PubMed] [Google Scholar]
  • 61.Wallace G, Jodele S, Howell J, et al. Vitamin D Deficiency and Survival in Children after Hematopoietic Stem Cell Transplant. Biol Blood Marrow Transplant. 2015;21(9):1627–1631. doi: 10.1016/j.bbmt.2015.06.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Hansson MEA, Norlin A-C, Omazic B, et al. Vitamin D Levels Affect Outcome in Pediatric Hematopoietic Stem Cell Transplantation. Biol Blood Marrow Transplant. 2014;20(10):1537–1543. doi: 10.1016/j.bbmt.2014.05.030 [DOI] [PubMed] [Google Scholar]
  • 63.Wolfrum S, Jensen KS, Liao JK. Endothelium-dependent effects of statins. Arterioscler Thromb Vasc Biol. 2003;23(5):729–736. doi: 10.1161/01.ATV.0000063385.12476.A7 [DOI] [PubMed] [Google Scholar]
  • 64.Zeisbrich M, Becker N, Benner A, et al. Transplant-associated thrombotic microangiopathy is an endothelial complication associated with refractoriness of acute GvHD. Bone Marrow Transplant. 2017;52:1399. 10.1038/bmt.2017.119. [DOI] [PubMed] [Google Scholar]
  • 65.Takatsuka H, Takemoto Y, Iwata N, et al. Oral eicosapentaenoic acid for complications of bone marrow transplantation. Bone Marrow Transplant. 2001;28(8):769–774. doi: 10.1038/sj.bmt.1703226 [DOI] [PubMed] [Google Scholar]
  • 66.Gomez SA, Abrey-Recalde MJ, Panek CA, et al. The oxidative stress induced in vivo by Shiga toxin-2 contributes to the pathogenicity of haemolytic uraemic syndrome. Clin Exp Immunol. 2013;173(3):463–472. doi: 10.1111/cei.12124 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Mezö B, Horváth O, Sinkovits G, Veszeli N, Kriván G, Prohászka Z. Validation of Early Increase in Complement Activation Marker sC5b-9 as a Predictive Biomarker for the Development of Thrombotic Microangiopathy After Stem Cell Transplantation. Front Med. 2020;7:646. https://www.frontiersin.org/article/10.3389/fmed.2020.569291. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES