4
views
0
recommends
+1 Recommend
2 collections
    0
    shares
      • Record: found
      • Abstract: not found
      • Article: not found

      Everolimus-associated alveolar hemorrage relapse after drug discontinuation in a kidney transplant recipient

      other

      Read this article at

      ScienceOpenPublisherPMC
      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

          The Case We report the case of a 51-year-old man affected by End Stage Renal Disease due to diabetic nephropathy, who received a kidney transplantation from a deceased donor. After induction with basiliximab and high dose intravenous methylprednisolone boluses, the patient was on maintenance immunosuppression with methylprednisolone, tacrolimus and everolimus. Everolimus was chosen in order to minimize calcineurin inhibitor levels in a patient with a long history of post-surgical diabetes mellitus. Major comorbidities and other medications are reported in the Supplementary Materials. Two months after transplantation, the patient started suffering from dyspnoea and haemoptysis, in the absence of other associated symptoms. He was conscious, hemodynamically stable and apyretic, and rhinopharyingeal swab for Sars-CoV-2 was negative. During this period, everolimus and tacrolimus trough levels always remained within the optimal ranges (3–8 ng/ml and 2–4 ng/ml, respectively). Chest auscultation revealed diffuse inspiratory crackles and expiratory wheezing in both lungs. Chest X-ray showed bilateral alveolar micronodules with interstitial thickening. Subsequently the patient underwent high resolution CT scan (HRCT) that showed bilateral multiple “ground-glass” areas and infiltrates consistent with alveolar haemorrhage (Fig. 1: day 1 (a), day 17 (b), day 20 (c) and at 2-month follow up (d)). Fig. 1 HRCT scan on day 1 (a), day 17 (b), day 20 (c) and at a 2-month follow up (d). Empiric antibiotic therapy with piperacillin/tazobactam was started though with no response. Five days later, blood tests showed severe anaemia (Hb 7.2 g/dL), renal function impairment (eGFR 21 ml/min vs 35 ml/min), with normal white blood cell and platelet counts and C-reactive protein levels. Everolimus was discontinued and the patient was admitted to the Nephrology Unit: arterial blood gas analysis showed severe respiratory insufficiency requiring high-flux oxygen therapy. A red blood cell transfusion was performed. Further blood tests documented negative ANA, ANCA and AntiGBM antibodies. Blood and sputum cultures were negative, as were Quantiferon TB and polymerase chain reaction for CMV, EBV and BKV DNA. Fibrobroncoscopy was performed and broncho-alveolar lavage (BAL) showed progressively more bloody samples, typical of diffuse alveolar haemorrhage, which tested negative for bacterial, viral and fungal cultures; cytological evaluation detected several hemosiderin-laden macrophages (>20%) (Fig. 2). Fig. 2 Bloody samples from broncho-alveolar lavage fluid During hospitalization his clinical picture progressively improved with normalization of gas exchanges. Follow-up HRCT revealed global regression of the pathologic findings, albeit without complete resolution. Considering the rapid response to everolimus discontinuation, once other infectious and immunological differential diagnoses were excluded, a diagnosis of everolimus-related non-infectious pneumonitis was made. Therapy with high-dose intravenous steroids was considered, but not prescribed due to the rapid improvement of the clinical picture. The patient was discharged under immunosuppression with methylprednisolone and tacrolimus. However, the patient was re-admitted ten days later due to a relapse of dyspnoea, haemoptysis and asthenia. New blood tests showed a relapse of severe anaemia and HRCT revealed worsening of the “ground-glass”-like areas and alveolar haemorrhage. Pulses of intravenous steroids the clinical picture progressively improved up to complete resolution of symptoms, that led to hospital discharge. Rapid tapering of steroids to the maintenance dose (predsnisone 5 mg daily) was carried out and currently, after 12 months of follow-up, no relapse has occurred. Clinical course is summarised in Fig. 3. Fig. 3 Timeline of clinical course and drug administration Lesson for the clinical nephrologist Adverse events observed in patients undergoing systemic treatment with mammalian target of rapamycin (mTOR) inhibitors include pulmonary dysfunction (non-infectious pneumonitis), epithelial-cutaneous events (stomatitis, rash), metabolic dysfunction (elevated blood levels of glucose and lipids), and immune suppression (infections). MTOR inhibitors are commonly used for cancer treatment and as immunosuppressants to prevent rejection of solid organ transplants. Everolimus, sirolimus and temsirolimus belong to this class of drugs and are generally well tolerated (1). The most common side effects related to everolimus include stomatitis, cutaneous rash, metabolic dysfunction, infections, oedema, proteinuria and difficult wound healing (2), while pulmonary toxicity is a rare complication that occurs in up to 16% of patients receiving this mTORi as chemotherapy and in 4.3% of transplant recipients receiving everolimus as an immunosuppressant (1, 3). The cause of this potentially serious adverse event is not yet fully understood. Rapamycin and its derivatives can induce autophagy in cells by inhibiting mTORC1, thus causing phosphorylation of pro-autophagic kinases and determining autophagosome formation and possible exposure of antigens on alveolar endothelial cells,which may explain a lymphocyte-mediated toxicity (4). On the other hand, the clinical response to corticosteroids and discontinuation of everolimus, together with T-cell lymphocytosis often reported in bronchoalveolar lavage, suggests a hypersensitivity mechanism (1). According to another hypothesis, the high affinity of sirolimus for plasma proteins may render it immunogenic as a hapten, thus triggering a T-cell reaction against the antigen complex (5). Direct toxicity may be suggested by the greater rates of pneumonitis occurring in patients receiving high doses of mTORi and by the fact that discontinuation of therapy is necessary to allow early remission of the symptoms (6, 7). Furthermore, pulmonary toxicity does not seem to be related to longer drug exposure and/or higher blood levels, since many reports of pneumonia are described in patients whose everolimus levels are within therapeutic range (4, 6). Once the diagnosis is made, clinical management largely depends on the severity of the symptoms. Light to moderate cases usually benefit from drug dose reduction or discontinuation and oral corticosteroid therapy (7), while the literature regarding a therapeutic shift from everolimus to another mTORi is controversial (7, 8). Patients with severe to potentially deadly clinical pictures showed good response to everolimus discontinuation and high-dose intravenous corticosteroid therapy, which should be administered while the evaluation is on-going if there is a solid clinical suspicion of non-infectious pneumonitis (1, 7). We report the case of a transplant recipient who developed severe respiratory insufficiency due to alveolar haemorrhage, confirmed by fibrobronchoscopy, under everolimus treatment. Bacteriological, mycobacteriological and viral tests allowed excluding the most common infectious aetiologies; auto-immune study was also negative. The clinical picture was not modified by broad spectrum antibiotic therapy, and indeed, it was responsive to everolimus discontinuation. After observing an initial rapid clinical resolution of symptoms and radiological signs of improvement, a relapse of the clinical picture occurred almost 20 days after the discontinuation of everolimus, which resolved only after intravenous pulse steroids. Relapse of symptoms after everolimus discontinuation might lead us to speculate that the first diagnosis was wrong, however, after this episode no other relapses were observed during 12 months of follow-up with standard low-dose steroid maintenance therapy. Non-infectious pneumonia related to everolimus is a rare and potentially harmful complication that must always be considered in the differential diagnosis in patients receiving mTOR inhibitors and complaining of non-specific respiratory symptoms, with or without systemic conditions like fever or asthenia. Accurate management of this class-related side effect is crucial, and the clinical approach must be tailored to the severity of the condition. Pulmonary function tests were not performed in the reported case. Of note, the use of diffusing capacity of the lung for carbon monoxide (DLCO) combined with HRCT scan has been proposed in an algorithm for surveillance of fragile patients (such as recipients of solid organ transplants), especially when more than one drug with known pulmonary potential toxicity is simultaneously administered (9). This is not the first reported case of everolimus-related pneumonitis in the literature, however, it may suggest paying attention to some peculiar aspects: the severity of presentation with alveolar haemorrhage and a late relapse after drug discontinuation. Improvement of symptoms should not be overestimated since relapse is possible, and the most severe cases, lithe the one here described, may require high dose intravenous steroid administration to achieve complete remission. Supplementary Information Below is the link to the electronic supplementary material. Supplementary file1 (DOCX 13 kb)

          Related collections

          Most cited references9

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

          AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1.

          Autophagy is a process by which components of the cell are degraded to maintain essential activity and viability in response to nutrient limitation. Extensive genetic studies have shown that the yeast ATG1 kinase has an essential role in autophagy induction. Furthermore, autophagy is promoted by AMP activated protein kinase (AMPK), which is a key energy sensor and regulates cellular metabolism to maintain energy homeostasis. Conversely, autophagy is inhibited by the mammalian target of rapamycin (mTOR), a central cell-growth regulator that integrates growth factor and nutrient signals. Here we demonstrate a molecular mechanism for regulation of the mammalian autophagy-initiating kinase Ulk1, a homologue of yeast ATG1. Under glucose starvation, AMPK promotes autophagy by directly activating Ulk1 through phosphorylation of Ser 317 and Ser 777. Under nutrient sufficiency, high mTOR activity prevents Ulk1 activation by phosphorylating Ulk1 Ser 757 and disrupting the interaction between Ulk1 and AMPK. This coordinated phosphorylation is important for Ulk1 in autophagy induction. Our study has revealed a signalling mechanism for Ulk1 regulation and autophagy induction in response to nutrient signalling.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Noninfectious pneumonitis after everolimus therapy for advanced renal cell carcinoma.

            Noninfectious pneumonitis is a known class effect of mammalian target of rapamycin (mTOR) inhibitors. To assess the incidence, radiographic patterns, management, and outcome of pneumonitis in patients with advanced renal cell carcinoma receiving everolimus. Clinical study data from 416 patients, randomized to receive everolimus versus placebo, were analyzed for adverse events of pneumonitis. Radiographic studies performed every 8 weeks were subject to a prospective, independent, blinded central review for the presence of findings indicative of pneumonitis. Of 274 patients receiving everolimus, clinical pneumonitis was suspected for 37 patients (13.5%) (none with placebo). Nine cases (3.3%) were grade 1 (asymptomatic), 18 (6.6%) were grade 2 (not interfering with daily living), and 10 (3.6%) were grade 3 (interfering with daily living or oxygen indicated). No grade 4 (life-threatening) pneumonitis was observed. Of the 10 patients with grade 3 pneumonitis, 5 had baseline radiological evidence of pneumonitis before everolimus therapy. Twenty of the 37 cases (54.0%) were reversible within the follow-up period; resolution followed dose reduction for 20 patients and treatment discontinuation in 10 patients. Corticosteroid therapy was initiated in 16 cases. Dedicated radiological review of available serial radiographic studies (245 patients receiving everolimus and 132 receiving placebo) found a higher percentage of new radiographic findings even in patients without a diagnosis of clinical pneumonitis who were receiving everolimus versus placebo (38.9 vs. 15.2%). Early recognition, prompt intervention, and a conservative approach are important in managing the risk associated with noninfectious pneumonitis in association with everolimus. Clinical trial registered with www.clinicaltrials.gov (NCT 00410124).
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Sirolimus-associated pulmonary toxicity.

              Pulmonary toxicity has recently been recognized as a potentially serious complication associated with sirolimus therapy. We further detail this condition on the basis of our own cases and those reported in the literature. We report three cases of suspected sirolimus-induced pulmonary toxicity that occurred in three renal transplant recipients and searched PubMed for all previously reported cases. Including our current cases, 43 patients with sirolimus-induced pulmonary toxicity have now been reported. Clinical data were incomplete in 28 cases. Analysis of available data for 15 patients revealed that the most commonly presenting symptoms were dyspnea on exertion and dry cough followed by fatigue and fever. Chest radiographs and high-resolution computed tomography scans commonly revealed bilateral patchy or diffuse alveolo-interstitial infiltrates. Bronchoalveolar fluid analysis and lung biopsy in selected case reports revealed several distinct histologic features, including lymphocytic alveolitis, lymphocytic interstitial pneumonitis, bronchoalveolar obliterans organizing pneumonia, focal fibrosis, pulmonary alveolar hemorrhage, or a combination thereof. The diagnosis of sirolimus-associated pulmonary toxicity was made after an exhaustive work-up to exclude infectious causes and other pulmonary disease. Sirolimus discontinuation or dose reduction resulted in clinical and radiologic improvement in all 15 patients within 3 weeks. The temporal relationship between sirolimus exposure and onset of pulmonary symptoms in the absence of infectious causes and other alternative pulmonary disease and the associated clinical and radiologic improvement after its cessation suggests a causal relationship. Because the use of sirolimus in organ transplantation has become more widespread, clinicians must remain vigilant to its potential pulmonary complication.
                Bookmark

                Author and article information

                Contributors
                marco.busutti@live.com
                Journal
                J Nephrol
                J Nephrol
                Journal of Nephrology
                Springer International Publishing (Cham )
                1121-8428
                1724-6059
                14 March 2022
                : 1-4
                Affiliations
                [1 ]GRID grid.412311.4, Nephrology, Dialysis and Transplant Unit, IRCCS Policlinico Sant’Orsola, , Azienda Ospedaliero-Universitaria di Bologna, ; Bologna, Italy
                [2 ]GRID grid.412311.4, Interventional Pulmonology Unit, IRCCS Policlinico Sant’Orsola, , Azienda Ospedaliero-Universitaria di Bologna, ; Bologna, Italy
                Author information
                http://orcid.org/0000-0002-3150-6704
                Article
                1298
                10.1007/s40620-022-01298-w
                8919162
                35286598
                1d421527-edcc-4216-b2f8-994df7586621
                © The Author(s) under exclusive licence to Italian Society of Nephrology 2022

                This article is made available via the PMC Open Access Subset for unrestricted research re-use and secondary analysis in any form or by any means with acknowledgement of the original source. These permissions are granted for the duration of the World Health Organization (WHO) declaration of COVID-19 as a global pandemic.

                History
                : 16 January 2022
                : 26 February 2022
                Categories
                Lessons for the Clinical Nephrologist

                kidney,lung,transplant,everolimus,toxicity
                kidney, lung, transplant, everolimus, toxicity

                Comments

                Comment on this article