13
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      An Update in Drug-Induced Thrombotic Microangiopathy

      discussion

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Introduction Drug-induced thrombotic microangiopathy (DITMA) is a life-threatening complication that is often under-recognized and under-reported (1). Despite recent systematic reviews published in 2015 (2) and 2018 (3), the list of drugs implicated in TMA continues to expand (4–9). In addition, novel reports have unraveled potential new mechanisms that might contribute to a targeted therapy of this syndrome. In this opinion article, we aimed to summarize recent data on DITMA, categorizing drugs based on mechanisms of actions and specialties. Mechanisms of Action Two decades ago, George et al. introduced the term “drug-induced thrombocytopenia.” Clinically based criteria were proposed and levels of evidence were stratified in order to solidify a definite, propable, possible, or an unlikely causal relationship between a drug and thrombocytopenia (10). Although the mechanisms of endothelial injury during DITMA still remain unknown; immune-mediated mechanisms or dose-dependent and cumulative toxicity are implied (11). The hypothesis is based on the observation of the timing of TMA occurrence, the pattern of disease, the exclusion of a better explanation thorough investigation. DITMA suspicion is ampilified by TMA resolution when the drug is withdrawn or recurrent endothelial injury during re-exposure to the drug. During the last decade, laboratory criteria have been added to support the causal relationship between a drug and TMA (2). Some examples of drugs in which antibody mediated DITMA has been confirmed with identification of drug-dependent antibodies to platelets or other cells as the pathophysiologic mechanism of TMA are quinine, oxaliplatin, and vancomycin (12). On the other hand, the dose-dependent and cumulative toxicity model seems to fit for opana's abuse, bevacizumab, levofloxacin, alemtuzumab, and interferon's cases of DITMA (9, 13–15). It is important to exclude any other diagnosis before attributing TMA to a drug. For example, in some cases such as these of ipilimumab, pazopanib, ustekinumab, and golimumab severe ADAMTS13 deficiency was found, plasma exchange was effective and no drug-dependent antibody inhibition of ADAMTS13 activity was reported, making drug-indused causal relationship unlikely (7, 16–18). Figure 1 summarizes postulated mechanisms in DITMA. Figure 1 Summary of postulated mechanisms in drug-induced thrombotic microangiopathy (DITMA). ADAMTS13: a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13. Hematology/Oncology DITMA is caused by various drugs used in Hematology and Oncology. Chemotherapy Chemotherapeutic agents were the first to be implicated in causing DITMA. Mitomycin and gemcitabine have numerous reports of dose-related DITMA, while one report describes an immune-DITMA as a result of gemcitabine administration (19–26). Despite their use as a combination with other drugs, which makes the direct causal relationship difficult in some cases, many well-described cases support a clear-cut association (21, 27, 28). Three reports have described DITMA caused by pentostatin, a purine analog used in lymphoproliferative diseases (29). Docetaxel and vincristine have also been reported to induce TMA (30, 31). Oxaliplatin has been implicated as a cause of DITMA in a review by Al-Nouri et al. (2), although the authors of the original report described gemcitabine as the causative factor (32). Renal-limited TMA has been reported in three patients treated with pegylated liposomal doxorubicin (33) and in one patient receiving treatment with a short interfering RNA targeted against Myc (DCR-MYC) (5). Multiagent Chemotherapy Drug-induced TMA has been reported in children with acute lymphoblastic leukemia (34, 35) and adults with solid tumors (36, 37), receiving multiagent chemotherapy. Jodele et al. described 13 patients developing TMA after high-dose chemotherapy and autologous stem cell transplantation for neuroblastoma (12 patients receiving carboplatin/etoposide/melphalan and one cyclophosphamide/thiotepa) (38). Finally, a high incidence of TMA was observed in melanoma patients receiving a lymphodepleting preparative chemotherapy regimen with total body irradiation (TBI) prior to autologous T cell therapy (39). In all cases, the co-administration of multiple drugs hinders the identification of the causative agent. Proteasome Inhibitors Proteasome inhibitors are mainly used in multiple myeloma treatment and have been associated with DITMA (bortezomib, carfilzomib, ixazomib). The majority of reports have implicated bortezomib and cafilzomib (3, 40). Recent reports also support a causal association of ixazomib with DITMA (41–43). Some authors report successful treatment of carfilzomib-induced TMA with eculizumab (44, 45). VEGF, Kinase and Immune Checkpoint Inhibitors Vascular endothelial growth factor (VEGF) inhibitors are used for the treatment of various malignancies. Many cases reported DITMA as a result of bevacizumab use, a humanized monoclonal antibody directed against VEGF (46–49). In some, treatment with eculizumab was successful (50). Ramucirumab, anti-VEGF receptor 2 monoclonal antibody, and cetuximab, a monoclonal antibody against epidermal growth factor receptor (EGFR), are also implicated in causing renal-limited TMA (51, 52). Tyrosine kinase inhibitors (TKIs) are effective in the treatment of hematologic malignancies and solid tumors. Sunitinib is a small-molecule TKI that targets VEGFR-2 and PDGFR-b. Imatinib and ponatinib are small-molecule BCR-ABL TKIs, used mainly in the treatment of chronic myeloid leukemia. Palbociclib inhibits the cyclin-dependent kinases CDK4 and CDK6. Cases reporting a link between the aforementioned TKIs and drug-induced TMA have been described in literature (6, 53–56). Two cases of TMA, one in a patient receiving the immune checkpoint inhibitor ipilimumab, and one in a patient treated with multi-targeted receptor tyrosine kinase inhibitor pazopanib have also been reported. However, these reports differ from other DITMAs, due to the severe ADAMTS13 deficiency (7, 16). Calcineurin and mTOR Inhibitors Numerous reports implicate cyclosporine and tacrolimus in causing dose-dependent TMA (2). Most of these reports described calcineurin inhibitor-induced TMA in patients that have undergone hematopoeitic stem cell or solid organ transplantation (57). Calcineurin inhibitor-induced TMA mostly affects the kidneys (58, 59). The inhibitors of the mechanistic target of rapamycin (mTOR), can also cause DITMA, most frequently associated with sirolimus or tacrolimus administration, than everolimus (60–62). Successful treatment with complement inhibition has been described in several patients, since this condition along with TA-TMA has been considered to resemble atypical hemolytic uremic syndrome (aHUS) (63, 64). Monoclonal Antibodies The first reported case of monoclonal antibody-induced TMA described a patient treated with anti-T cell monoclonal antibody muromonab-CD3 (OKT3) (65). Emicizumab, a monoclonal antibody used in Hemophilia A, co-administered with high doses of activated prothrombin complex concentrate (aPCC) has been linked with TMA in three patients (66). Discontinuation of aPCC resulted in resolution of TMA, highlighting the fact that emicizumab monotherapy may not be sufficient to cause DITMA. Various monoclonal antibodies against tumor necrosis factor alpha (TNF-a), such as adalimumab, golimumab, and certolizumab pegol have been reported to cause TMA in a few cases (17, 18, 67). Another report describes a patient with psoriasis developing TMA after treatment with methotrexate and ustekinumab, a monoclonal antibody that blocks interleukin (IL)-12 and IL-23 (68). However, in the cases where ustekinumab and golimumab were suspected to be the causative factor of DITMA, the authors reported low levels of ADAMS13 (<5%) and an initial response to plasma exchange, making DITMA diagnosis unlikely (17, 18). In phase 1 study, moxetumomab pasudotox, an anti-CD22 immunotoxin used in the treatment of childhood acute lymphoblastic leukemia, caused TMA in 13% of patients. In the majority of cases, TMA resolved with drug discontinuation (69). Opioids and Other Drugs of Abuse Intravenous use of the extended-release opioid oxymorphone and oxycodone tablets reformulated with polyethylene oxide (PEO) have been reported to cause DITMA in many patients (3). Subsequently, intravenously administrated high molecular PEO was determined as the causative factor (15). Cocaine and ecstasy have caused DITMA in recreational users (70–72). Neurology One of the biggest challenges in neurology is the lack of disease-specific drugs that contributes to the increasing global burden of neurological disorders (73). Traditionally, epilepsy benefited from a wide variety of available medicines but during the last decade numerus drugs were introduced at multiple sclerosis (MS) treatment raising long-term safety considerations (74, 75). Until 2018, interferon beta 1-a and 1-b, disease modifying treatments (DMTs) of MS and anticonvulsive valproic acid were the only neurologic drugs associated with thrombotic microangiopathy (3). Recently, alemtuzumab; which was approved by US Food and Drug Administration (FDA) for treatment of relapsing-remitting MS (RRMS) at 2014; was associated for the first time with DITMA (8, 9). Administration of alemtuzumab was known to rarely cause severe renal adverse effects (76). Nevertheless, in that case report the causal relationship of alemtuzumab with TMA is supported by the fact that (a) symptoms started immediately after the first infusion and (b) the patient did not respond to plasma exchange (9). Another DMT, fingolimod was linked with TMA in an induced-malignant hypertension animal model; in contrast with control group in which fingolimod was not administrated (77). Interferon (IFN) has also been correlated with TMA with a dose-dependent manner (14). Further studies confirm that TMA is a severe complication of IFN-beta RRMS treatment. Lately, five patients were reported to have IFN-induced TMA following long-term treatment (78–80). Interestingly, renal function of three patients improved only after administration of eculizumab, not after withdrawal of IFN (80). Clinical translation of those studies raises awareness of neurologists for early recognition and management of TMA when prescribing DMTs. Infectious Diseases An infection can be caused by a variety of organisms such as bacteria, viruses, parasites or fungi. Many anti-infectives agents have been associated with DITMA in the past (2); quinine, the treatment of malaria, was the most commonly reported (81). Novel studies implicate a number of different drugs in causing DITMA. First of all, a case report incriminates hydroxychloroquine, a synthetic derivative of quinine used for rheumatoid arthritis and systemic lupus erythematosus, as a possible cause of thrombotic thrombocytopenia purpura (TTP) (82). Disease progression was detrimental and patient died in spite of drug withdrawal and plasma exchange. Moreover, for the first time an antiretroviral treatment of human immunodeficiency virus consisting of tenofovir/emtricitabine was found to have a causality relationship with immune TTP (83). After the cessation of the drug and the initiation of corticosteroids and azathioprine the patient recovered. Last but not least, several antibiotics such as ciprofloxacin, penicillin, and metronidazole were reported with probable evidence to cause DITMA (2). A new case report implicates again ciprofloxacin in drug-induced TTP which resolved completely with plasma exchange (84). Another report, identified a highly effective and frequently prescribed fluoroquinolone, levofloxacin as a new potential suspect for DITMA (13). This case report described two patients who developed microangiopathic hemolysis and thrombocytopenia following levofloxacin treatment of respiratory tract infections. Both cases resolved after drug cessation; the first patient received also therapeutic plasma exchange. In conclusion, a wide variety of anti-infectives agents have been scarcely correlated with DITMA and unfortunately, no one could predict or prevent its appearance; hence, it is of paramount importance to be aware of that possibility in order to start the appropriate treatment promptly. Therapeutic Potentials The only proven intervention in the management of DITMA is discontinuation of the offending agent. Plasma exchange and immunosuppressive therapy may be a reasonable treatment option, especially when the diagnosis is uncertain. Although rarely described in DITMA, patients with severe ADAMTS13 deficiency respond to plasma exchange (7, 16–18, 82–84). However, these reports should be interpreted with caution, since severe ADAMTS13 deficiency indicates TTP as a more likely diagnosis. In true DITMA, plasma exchange is ineffective (85). On the other hand, numerous reports have now confirmed that complement inhibition with eculizumab shows efficacy in DITMA (22, 26, 44, 45, 50, 63, 80). Eculizumab is a first-in-class monoclonal antibody that blocks terminal complement activation with proven safety and efficacy in complement-mediated TMAs (86). Based on current literature, we would consider eculizumab administration in three instances: in patients with non-immune DITMA, in those who deteriorate despite discontinuation of the implicated drug and supportive care, and finally, in patients at risk of kidney failure (87). Conclusions and Future Perspectives Our state-of-the-art report categorizes drugs that have been associated with DITMA, summarized in Table 1. It also emphasizes on unique presentations and characteristics of DITMA, that require increased awareness by treating physicians of relevant specialties. Hematologists are largely involved in the administration of the majority of these drugs, along with other internal medicine specialties. Since many patients have presented with renal-limited complications, the role of nephrologists is also important. Therefore, our report highlights an unmet clinical need of increased recognition and better understanding of DITMA by treating physicians across different specialties. Table 1 Summary of drugs involved in DITMA. Drug Type Specialty Docetaxel Doxorubicin DCR-MYC Gemcitabine Oxaliplatin Pentostatin Vincristine Chemotherapy Hematology/Oncology Carboplatin + Etoposide + Melphalan Cyclophosphamide + Thiotepa Multiagent chemotherapy Bortezomib Carfilzomib Ixazomib Proteasome inhibitors Bevacizumab Ramucirumab Cetuximab Imatinib Ipilimumab Pazopanib Ponatinib Palbociclib Ruxolitinib Sunitinib VEGF, kinase and immune checkpoint inhibitors Cyclosporine Rapamycin Tacrolimus Calcineurin and mTOR inhibitors Adalimumab Certolizumab pegol Emicizumab + aPCC Golimumab OKT3 Ustekinumab Moxetumomab pasudotox Monoclonal antibodies Hematology/Oncology/ Rheumatology Cocaine/ Ecstasy Oxymorphone/Oxycodone Polyethylene oxide (PEO) Opioids / Drugs of abuse Toxicology Interferon beta 1-a /1-b Alemtuzumab Fingolimod Disease modifying treatments for Multiple Sclerosis Neurology Valproic acid Anticonvulsive Tenofovir/Emtricitabine Anti-infectives Infectious diseases Quinine/Hydroxychloroquine Antimalarials Ciprofloxacin Fluoroquinolone Metronidazole Penicillin Antibiotics Except for expanding the list of drugs associated with DITMA, future reports need to take into account potential mechanisms. Identification of underlying etiology is of utmost importance for proper management. Further mechanistic studies need to identify the drugs or pathways involved in complement activation in order to early select patients that would benefit from complement inhibition. Author Contributions EG conceived the manuscript concept. TC and MG wrote the manuscript. AA and EG edited and approved the final manuscript. Conflict of Interest The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

          Related collections

          Most cited references78

          • Record: found
          • Abstract: found
          • Article: not found

          Guidelines on the Use of Therapeutic Apheresis in Clinical Practice-Evidence-Based Approach from the Writing Committee of the American Society for Apheresis: The Seventh Special Issue.

          The American Society for Apheresis (ASFA) Journal of Clinical Apheresis (JCA) Special Issue Writing Committee is charged with reviewing, updating, and categorizing indications for the evidence-based use of therapeutic apheresis in human disease. Since the 2007 JCA Special Issue (Fourth Edition), the Committee has incorporated systematic review and evidence-based approaches in the grading and categorization of apheresis indications. This Seventh Edition of the JCA Special Issue continues to maintain this methodology and rigor to make recommendations on the use of apheresis in a wide variety of diseases/conditions. The JCA Seventh Edition, like its predecessor, has consistently applied the category and grading system definitions in the fact sheets. The general layout and concept of a fact sheet that was used since the fourth edition has largely been maintained in this edition. Each fact sheet succinctly summarizes the evidence for the use of therapeutic apheresis in a specific disease entity. The Seventh Edition discusses 87 fact sheets (14 new fact sheets since the Sixth Edition) for therapeutic apheresis diseases and medical conditions, with 179 indications, which are separately graded and categorized within the listed fact sheets. Several diseases that are Category IV which have been described in detail in previous editions and do not have significant new evidence since the last publication are summarized in a separate table. The Seventh Edition of the JCA Special Issue serves as a key resource that guides the utilization of therapeutic apheresis in the treatment of human disease. J. Clin. Apheresis 31:149-162, 2016. © 2016 Wiley Periodicals, Inc.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Drug-induced thrombocytopenia: a systematic review of published case reports.

            To determine the strength of clinical evidence for individual drugs as a cause of thrombocytopenia. All English-language reports on drug-induced thrombocytopenia. Articles describing thrombocytopenia caused by heparin were excluded from review. Of the 581 articles reviewed, 20 were excluded because they contained no patient case reports. The remaining 561 articles reported on 774 patients. Two of the authors used a priori criteria to independently review each patient case report. Two hundred fifty-nine patient case reports were excluded from further review because of lack of evaluable data, platelet count of 100000 cells/microL or more, use of cytotoxic or nontherapeutic agents, occurrence of drug-induced systemic disease, or occurrence of disease in children. For the remaining 515 patient case reports, a level of evidence for the drug as the cause of thrombocytopenia was assigned. Data on bleeding complications and clinical course were recorded. The evidence supported a definite or probable causal role for the drug in 247 patient case reports (48%). Among the 98 drugs described in these reports, quinidine was mentioned in 38 case reports, gold in 11, and trimethoprim-sulfamethoxazole in 10. Of the 247 patients described in the case reports, 23 (9%) had major bleeding and 2 (0.8%) died of bleeding. Many reports of drug-induced thrombocytopenia do not provide evidence supporting a definite or probable causal relation between the disease and the drug. Future patient case reports should incorporate standard criteria to clearly establish the etiologic role of the drug.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Complementopathies and precision medicine

              The renaissance of complement diagnostics and therapeutics has introduced precision medicine into a widened field of complement-mediated diseases. In particular, complement-mediated diseases (or complementopathies) with ongoing or published clinical trials of complement inhibitors include paroxysmal nocturnal hemoglobinuria, cold agglutinin disease, hemolytic uremic syndrome, nephropathies, HELLP syndrome, transplant-associated thrombotic microangiopathy, antiphospholipid antibody syndrome, myasthenia gravis, and neuromyelitis optica. Recognizing that this field is rapidly expanding, we aim to provide a state-of-the-art review of (a) current understanding of complement biology for the clinician, (b) novel insights into complement with potential applicability to clinical practice, (c) complement in disease across various disciplines (hematology, nephrology, obstetrics, transplantation, rheumatology, and neurology), and (d) the potential future of precision medicine. Better understanding of complement diagnostics and therapeutics will not only facilitate physicians treating patients in clinical practice but also provide the basis for future research toward precision medicine in this field.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Med (Lausanne)
                Front Med (Lausanne)
                Front. Med.
                Frontiers in Medicine
                Frontiers Media S.A.
                2296-858X
                22 May 2020
                2020
                : 7
                : 212
                Affiliations
                [1] 1BMT Unit, Hematology Department, G Papanicolaou Hospital , Thessaloniki, Greece
                [2] 2Laboratory of Clinical Neurophysiology, AHEPA Hospital, Aristotle University of Thessaloniki , Thessaloniki, Greece
                Author notes

                Edited by: Robert W. Maitta, Case Western Reserve University, United States

                Reviewed by: Sonata Jodele, Cincinnati Children's Hospital Medical Center, United States

                *Correspondence: Eleni Gavriilaki elenicelli@ 123456yahoo.gr

                This article was submitted to Hematology, a section of the journal Frontiers in Medicine

                †These authors have contributed equally to this work

                Article
                10.3389/fmed.2020.00212
                7256484
                32528969
                58538211-a664-482f-9513-cc4644950254
                Copyright © 2020 Chatzikonstantinou, Gavriilaki, Anagnostopoulos and Gavriilaki.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 12 February 2020
                : 29 April 2020
                Page count
                Figures: 1, Tables: 1, Equations: 0, References: 87, Pages: 7, Words: 4933
                Categories
                Medicine
                Opinion

                thrombotic microangiopathy,drug,hematology,oncology,neurology

                Comments

                Comment on this article