mycoses

Diagnosis,Therapy and Prophylaxis of Fungal Diseases

Review article

Our 2015 approach to invasive pulmonary aspergillosis € nke,5 G. Michels,5 B. Liss,1,2 J. J. Vehreschild,1,2,3 C. Bangard,4 D. Maintz,4 K. Frank,5 S. Gro A. Hamprecht,6 H. Wisplinghoff,6 B. Markiefka,7 K. Hekmat,8 M. J. G. T. Vehreschild1,2,3 and O. A. Cornely1,2,3,9,10 Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany, 2Center for Integrated Oncology CIO K€ olnBonn, University of Cologne, Cologne, Germany, 3German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Cologne, Germany, 4Department of Radiology, University Hospital of Cologne, Cologne, Germany, 5Department III of Internal Medicine, Heart Centre of the University of Cologne, Cologne, Germany, 6 Institute for Medical Microbiology, Immunology and Hygiene, University Hospital of Cologne, Cologne, Germany, 7Institute of Pathology, University Hospital of Cologne, Cologne, Germany, 8Department of Cardiothoracic Surgery, University Hospital of Cologne, Cologne, Germany, 9Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany and 10Clinical Trials Centre Cologne, ZKS K€oln, University of Cologne, Cologne, Germany 1

Summary

At the University Hospital of Cologne, in general two patient groups at high risk for invasive aspergillosis receive posaconazole prophylaxis: Acute myelogenous leukaemia patients during remission induction chemotherapy and allogeneic haematopoietic stem cell transplant recipients. Other patients at risk undergo serum galactomannan testing three times weekly. At 72–96 h of persisting fever despite broad-spectrum antibiotics, or at onset of lower respiratory tract symptoms a thoracic computed tomography (CT) scan is performed. Without lung infiltrates on CT, IPA is ruled out. In lung infiltrates not suggestive for IPA mycological confirmation is pursued. In patients without posaconazole prophylaxis empiric caspofungin will be considered. CT findings typical for IPA prompt targeted treatment, and mycological confirmation. Bronchoalveolar lavage (BAL) is most important for cultural identification and susceptibility testing, and facilitates diagnosing other pathogens. BAL performance is virtually independent of platelet counts. If despite suggestive infiltrates BAL does not yield the diagnosis, CT-guided biopsy follows as soon as platelet counts allow. Surgery can also be beneficial in diagnosis and treatment of IPA. If the diagnosis of IPA is not established, mucormycosis is a valid concern. In patients with breakthrough IPA during posaconazole prophylaxis liposomal amphotericin B is the drug of choice. If no posaconazole prophylaxis was given, voriconazole is the treatment of choice for IPA.

Key words: Aspergillosis, mould pneumonia, neutropaenia, voriconazole, liposomal amphotericin B, caspofungin.

Introduction Current guidelines and recommendations of professional societies are important sources of information. As they might not always be applicable to specific clinical Correspondence: Prof. O. A. Cornely, University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany. Tel.: +49 221 478 6494. Fax: +49 221 478 3611. E-mail: [email protected] Submitted for publication 5 March 2015 Revised 9 March 2015 Accepted for publication 9 March 2015

© 2015 Blackwell Verlag GmbH

settings, we describe our institutional standard clinical approach to invasive pulmonary aspergillosis.1–4 Invasive pulmonary aspergillosis is a major challenge in the management of immunocompromised patients, due to a unique combination of high attributable mortality of ~70% and notorious diagnostic difficulties.5 Incidence rates depend on the underlying disease and on treatments received. In solid organ transplant recipients incidence rates are 1.8–2.7%,6,7 and in haematological malignancy range from 4.7 to 13.1%.5 Despite protection of patients with long-term neutropaenia receiving posaconazole prophylaxis, in general the incidence of invasive aspergillosis is increasing.4,8–10 This

doi:10.1111/myc.12319

B. Liss et al.

development may be due to rising patient numbers in vulnerable populations,11 as well as more frequent use of aggressive therapies.12 Patients in intensive care units (ICU) apparently are at increased risk, but this is not yet understood, owing to the heterogeneity of their underlying diseases and the lack of a definition of ICU itself. Susceptibility in any of these patient groups is enhanced by factors as diverse as use of glucocorticosteroids and other immunosuppressants,13 preexisting structural lung damage14–16 and genetic predisposition.17,18 Timely treatment reduces mortality rates,19 but is often hampered by a lack of specific clinical signs and symptoms,20 and by unreliable in vitro diagnostic assays.21 A standardised approach may increase physician compliance and thus diagnostic yield allowing earlier targeted treatment and subsequently lead to improved outcome.22–24 At our university hospital, we follow standard operating procedures (SOP) for diagnosis and treatment of infections in neutropaenic hosts. In an additional poster format the ‘febrile neutropaenia SOP’ are readily available on our haematology and stem cell transplant wards. In addition, they are web-based 25 (Fig. 1 and Fig. 2) for easy reference. They follow published evidence and guideline documents of professional societies, e.g. ECMM, ESCMID, DGHO and are frequently updated.1–4

Diagnostic strategy Patients with acute myelogenous leukaemia, myelodysplastic syndrome (AML/MDS) or severe aplastic anaemia and recipients of allogeneic haematopoietic stem cell transplantation receive posaconazole prophylaxis at our institution.4,9,10,26 Posaconazole prophylaxis has been included into the appropriate chemotherapy regimes in our ordering software. Patients ineligible for azole prophylaxis, e.g. those with acute lymphoblastic leukaemia, undergo serum galactomannan screening three times weekly.27,28 The most prevalent, but unspecific sign of invasive pulmonary aspergillosis, is persistent fever despite broadspectrum antibiotic treatment, reported in up to 100% of patients.20,29 A computer tomographic scan is ordered after 72–96 h of persistent fever and is performed within 24 h, usually on the same day. This thoracic CT scan without contrast enhancement is a rapid procedure, thus not interfering with planned examinations in our radiology department. In any patient with lower respiratory tract symptoms a CT scan is ordered independent of the presence and duration of fever.30 Diagnostic imaging is of utmost importance as all further diagnostic and therapeutic decisions depend on CT findings. We broadly categorise into no infiltrate, infiltrate not suggestive of invasive pulmonary aspergillosis, and infiltrate suggestive of invasive pulmonary aspergillosis, i.e.

Chest CT without contrast

Infiltrate suggestive of fungal infection (nodule, halo, cavity, air-crescent)

Unspecific infiltrate

No infiltrate

Invasive mold infection likely Bronchoalveolar lavage – specific test: Galactomannan, PCP Respiratory viruses Bacteria (including TBC)

High probability of invasive aspergillosis

Yes

Microscopy with septate hyphae or galactomannan (≥0.5 BAL or 2x in serum)

Other pathogen identified

No

Targeted aspergillosis therapy

Targeted therapy

No pathogen identified

No targeted therapy

Figure 1 Diagnostic algorithm for persistently febrile patients at risk of invasive pulmonary aspergillosis.

2

© 2015 Blackwell Verlag GmbH

Approach to invasive pulmonary aspergillosis

Posaconazole prophylaxis received? No

Yes

Liposomal amphotericin B 3 mg/kg QD i.v.

Voriconazole 6 mg/kg BID d1 4 mg/kg BID i.v.

Caspofungin d1 70 mg QD 50 mg QD i.v.

CT at d7 and d14 Respone/stable

Progression at 14 d

Figure 4 Invasive pulmonary aspergillosis in a patient with Consider oral treatment after at least 7 d i.v. treatment

AML: Infiltrates in both lungs with ‘air-crescent sign’ in the largest infiltrate. Consider biopsy

Figure 2 Therapeutic algorithm for patient with high probability

of invasive pulmonary aspergillosis.

Figure 3 Invasive pulmonary aspergillosis in a patient with mul-

tiple myeloma: Two spiculated infiltrates in the left upper lobe, each with surrounding halo.

• •

Nodular shape, with or without halo (Fig. 3), Cavity within area of consolidation or ‘air-crescent’ (Fig. 4). A reversed halo sign demands immediate attention, too. Suggestive of mucormycosis it is beyond the topic of this perspective article and has been dealt with elsewhere.3,31,32

Decision tree guided by CT findings Persistent fever without infiltrate

This is the case in 34.5% of our patients at risk (Fig. 5). Invasive pulmonary aspergillosis is ruled out. If the patient received posaconazole prophylaxis, this

© 2015 Blackwell Verlag GmbH

will be continued. No empiric antifungals are initiated and this approach is safe and effective.33 In this clinical situation any single diagnostic test will have a low yield, but identifying the cause of fever prevents polypragmatic treatment. In the absence of local symptoms during immunosuppression imaging of the paranasal sinuses rules out sinusitis. Repeated blood cultures can help identify blood stream infection by difficult to culture organisms or, e.g. Candida spp.34 Posaconazole serum concentration may be determined if the oral suspension is used, although no uncontroversial target level has been defined.35–38 Serum concentration is difficult to predict despite the use of complex models identifying correlating factors.39,40 Use of posaconazole tablets renders therapeutic drug monitoring generally obsolete.41 If the patient was not receiving posaconazole prophylaxis, empiric antifungal treatment is considered.42,43 Additional blood cultures are mandatory before the empiric antibiotic regimen may be switched,43 and abdominal infection should be considered.44,45 MRI can help identify aspergillosis of bones and joints.46 If fever of unknown origin persists, thoracic CT scans will be repeated weekly. Infiltrate non-suggestive of invasive pulmonary aspergillosis

This situation is encountered in 36.6% of our patients (Fig. 5). Invasive pulmonary aspergillosis is possible and needs to be ruled out. If galactomannan screening results were negative, or not done, e.g. in the case of posaconazole prophylaxis, bronchoalveolar lavage (BAL) is indicated. While we test BAL fluid for bacterial and viral infection, we in particular aim at

3

B. Liss et al.

28.9% 34.5%

Persistent fever without infiltrate Infiltrate non-suggestive of invasive pulmonary aspergillosis Infiltrate suggestive of invasive pulmonary aspergillosis

36.6%

Figure 5 Distribution of CT findings in

persistently febrile patients at risk for IPA.

galactomannan and fungal culture to confirm presence of Aspergillus spp.47–49 We only initiate antifungal treatment after mycological confirmation, although empiric treatment is an option in patients who did not receive posaconazole prophylaxis.42 We repeat thoracic CT imaging weekly if no diagnosis established despite all the efforts described above. Infiltrate suggestive of invasive pulmonary aspergillosis

In 28.9% of our patient population such infiltrates are found (Fig. 5). Invasive aspergillosis is highly likely, but not proven. Due to our early CT imaging policy, we usually find early CT signs. So the majority of these infiltrates are nodular and with or without characteristic halo sign. Air-crescent signs develop later in the course of aspergillosis.23,50 Actually, we rarely see cavities within areas of consolidation or air-crescent signs even in follow-up CT imaging of invasive pulmonary aspergillosis. We do not know whether this is due to early detection and treatment. In high-risk patients we still complete the diagnostic work-up to facilitate targeted treatment, but initiate treatment independent of further confirmation.51 If no galactomannan or cultural proof of aspergillosis on bronchoalveolar lavage fluid is established, mucormycosis remains a concern that might drive antifungal choice.31,52 CT-guided percutaneous biopsy

CT-guided biopsy has a high diagnostic yield in patients with haematological malignancies.53 With every CT scan revealing new infiltrates of unknown aetiology, we discuss diagnostic options with the radiologist. It is

4

important that biopsies undergo both, histological and cultural evaluation. Only fungal culture warrants reliable pathogen identification and in addition allows susceptibility testing. To sample enough material for histological and cultural evaluation we perform true-cut CT-guided coaxial biopsies. A 17,5 gauge coaxial needle is introduced into the nodule or the consolidating infiltrate, the trocar is removed, the semi-automatic biopsy needle is introduced, and five to ten 18 gauge, 2 cm long true-cut biopsies are taken. The first samples are for microbiological work-up and the last ones are fixed in formalin for histological examination. We avoid transgression of emphysematous lung parenchyma and lung fissures. If necessary, platelets are transfused one concentrate right before and one during the intervention, to ensure a platelet count of 50 000 ll 1. To lose no time (and platelets) that count is not verified prior intervention. To ensure maximum diagnostic yield all clinical samples are examined by microscopy, microbiological culture and histopathology analysis. For culture, specimen are inoculated on non-selective agars, enrichment broth and agars selective for fungi (Sabouraud) and incubated at 30 and 37°C for 7–14 days. Histological samples are processed and evaluated in a predefined manner. Necessary staining, haematoxylin – eosin, periodic acid-Schiff and silver stain as well as microscopic features to be evaluated, i.e. general morphology, width, branching angle and septation of hyphae are standardised. Treatment

For the treatment of invasive pulmonary aspergillosis antifungal choice depends on prior posaconazole

© 2015 Blackwell Verlag GmbH

Approach to invasive pulmonary aspergillosis

prophylaxis, as we prefer to switch antifungal class. If no posaconazole prophylaxis was given, voriconazole1,54 is our primary choice owing to superior response rates and lower toxicity compared to conventional amphotericin B deoxycholate.55,56 Isavuconazole may find its place in this indication.57 If voriconazole is used, therapeutic drug monitoring is considered, as serum levels can vary considerably.58 However, neither target levels nor specifics of clinical implementation been firmly established. In the rare cases of breakthrough infection during posaconazole prophylaxis, we choose liposomal amphotericin B.1,54,59 Liposomal amphotericin B efficacy appears unhampered by prior azole exposure, in particular fluconazole, itraconazole and voriconazole, and clinically we extrapolate these findings to posaconazole.60 If both compounds are contraindicated, e.g. in patients with substantially elevated liver function tests or renal impairment, caspofungin is a reasonable alternative.61 Further treatment decisions build on clinical, mycological and radiological response. Despite adequate treatment, radiological findings may progress in the early phase of the disease, and several authors have reported peak volumes after 7 days of treatment, followed by a slow decline.50,62,63 In our experience, CT infiltrates should stabilise within 14 days of treatment initiation. Furthermore, air-crescent signs or cavities may become apparent, which are predictive of treatment success.62,63 For these reasons, we always perform CT scans after 7 and 14 days of treatment, allowing us to evaluate treatment success in awareness of the expected peak volume at day 7. Any progression of lung infiltrates in absence of air signs beyond 2 weeks of treatment is suggestive of treatment failure and associated with poor outcome. In this situation, renewed diagnostic efforts, especially biopsies for histological and cultural evaluation, may be lifesaving. The role of surgery

Many of our patients will undergo subsequent periods of immunosuppression, specifically neutropaenia. Knowing the cause of previous infections allows estimating the risk of these subsequent procedures. Open lung biopsy using video-assisted techniques has been advocated for diagnosing aspergillosis and differentiating other causes of pulmonary infiltrates, if submitted for histopathology and microbiology.64 Surgery is effective in the treatment of aspergillosis and their complications,65 as well as in preventing relapse

© 2015 Blackwell Verlag GmbH

of fungal infections in patients undergoing HSCT.66 The current guideline of the Infectious Diseases Society of America (IDSA) supports surgery for histological diagnostics, removal of residual infiltrates prior to the next chemotherapy cycle, acute haemoptysis, prevention of bleeding in the case of fungal lesions with vessel involvement and reduction in fungal burden.67 In recent years, we actually reintroduced surgery into our algorithm and regularly discuss surgical intervention to diagnose infection or to reduce fungal burden. As a rule, optimal timing in the individual patient may be immediate surgery during neutropaenia in case of mucormycosis, while we prefer surgical resection after resolution of neutropaenia and thrombocytopaenia in invasive pulmonary aspergillosis. Unmet medical needs

Newer non-invasive diagnostic tools emerge, but await further validation and standardisation before integrated into clinical practice pathways. Detection of fungal nucleic acid in human specimens is currently under assessment by the EORTC/MSG consensus group.68 While some PCR-based methods appear especially promising not only in diagnosing aspergillosis but also in concurrently assessing resistance.69 Lateral flow devices have been helpful in some studies in diagnosing invasive aspergillosis in conjunction with imaging studies.70,71 1,3-b-D-glucan (BDG) is increasingly used to diagnose invasive fungal infections and has also been proposed as a marker to guide treatment.72 While BDG is listed among the criteria for probable invasive fungal disease its role is unclear in the haematological patient populations, where false positive results have been reported.73–76 Recently, fungal-specific T-cell response has been found useful in diagnosing invasive mould infection, but validation in independent cohorts is pending.13,77–80 Further areas requiring investigation and not covered by current guidelines include increasing azole resistance. Though the SEPIA study (unpublished data and 81) and others find azole resistance at low rates,82 high numbers of azole-resistant isolates in a similar population have been reported,83 too. Also as of March 2015 no well-designed trials concerning the treatment of breakthrough infection have been published or registered at EudraCT or clinicaltrials.gov, possibly due to the rarity of these events.33,84–87 Finally, the benefit of antifungal combination treatment remains a matter of debate, despite increasing utilisation and promising results in certain subgroups.88,89

5

B. Liss et al.

Conclusion Whether institutional guidelines are worth the effort depends on physician compliance. Maintaining their usefulness is particularly challenging in diseases that demand interdisciplinary concerted action. We are convinced of the value of institutional guidelines, informed by international consensus recommendations and supplemented by recent evidence from clinical studies. Local epidemiology needs to be explored and incorporated into clinical pathways. Such institutional guidance expedites diagnosis, enables treatment customised to the individual patient, and finally improves outcome.

Conflict of interest BL, CB, DM, KF, SG, AH, HW, BM, KH have nothing to disclose. JJV is supported by the German Federal Ministry of Research and Education (BMBF grant 01KI0771) and the German Centre for Infection Research. JJV has received research grants from Astellas, Gilead Sciences, Infectopharm, Merck, Pfizer and Essex/ScheringPlough; and served on the speakers’ bureau of Astellas, Merck Sharp Dohme/Merck, Gilead Sciences, Pfizer and Essex/Schering-Plough. GM reports personal fees from Novartis and Berlin-Chemie AG, outside the submitted work. MJGTV has served at the speakers’ bureau of Pfizer, Merck, Gilead Sciences and Astellas Pharma, received research funding from 3M, Astellas Pharma and Gilead Sciences and is a consultant to Berlin Chemie. OAC is supported by the German Federal Ministry of Research and Education (BMBF grant 01KN1106), has received research grants from 3M, Actelion, Astellas, Basilea, Bayer, Celgene, Cubist/Optimer, Genzyme, Gilead, GSK, Merck/MSD, Miltenyi, Pfizer, Quintiles, Shionogi, Viropharma, is a consultant to 3M, Astellas, Basilea, Cidara, Cubist/Optimer, Da Volterra, Daiichi Sankyo, F2G, Genentech, Gilead, GSK, Merck/MSD, Merck Serono, Pfizer, Rempex, Sanofi Pasteur and Summit/Vifor, and received lecture honoraria from Astellas, Gilead, Merck/MSD and Pfizer.

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18 19

References 1

2

3

6

Bohme A, Ruhnke M, Buchheidt D et al. Treatment of invasive fungal infections in cancer patients–recommendations of the Infectious Diseases Working Party (AGIHO) of the German Society of Hematology and Oncology (DGHO). Ann Hematol 2009; 88: 97–110. Lagrou K, Buchheidt D, Richardson M et al. Diagnostic Group 2014 ESCMID Aspergillus GuidelineESCMID. Barcelona, Spain: EFSIG, 2014. Cornely OA, Cuenca-Estrella M, Meis JF, Ullmann AJ. European Society of Clinical Microbiology and Infectious Diseases (ESCMID) Fungal Infection Study Group (EFISG) and European Confederation of

20

21

22

23

Medical Mycology (ECMM) 2013 joint guidelines on diagnosis and management of rare and emerging fungal diseases. Clin Microbiol Infect 2014; 20(Suppl 3): 1–4. Tacke D, Buchheidt D, Karthaus M et al. Primary prophylaxis of invasive fungal infections in patients with haematologic malignancies. 2014 update of the recommendations of the Infectious Diseases Working Party of the German Society for Haematology and Oncology. Ann Hematol 2014; 93: 1449–56. Tsitsikas DA, Morin A, Araf S et al. Impact of the revised (2008) EORTC/MSG definitions for invasive fungal disease on the rates of diagnosis of invasive aspergillosis. Med Mycol 2012; 50: 538–42. Bodro M, Sabe N, Gomila A et al. Risk factors, clinical characteristics, and outcomes of invasive fungal infections in solid organ transplant recipients. Transplant Proc 2012; 44: 2682–5. Sganga G, Bianco G, Frongillo F, Lirosi MC, Nure E, Agnes S. Fungal infections after liver transplantation: incidence and outcome. Transplant Proc 2014; 46: 2314–8. Bitar D, Lortholary O, Le Strat Y et al. Population-based analysis of invasive fungal infections, France, 2001-2010. Emerg Infect Dis 2014; 20: 1149–55. Cornely OA, Maertens J, Winston DJ et al. Posaconazole vs. fluconazole or itraconazole prophylaxis in patients with neutropenia. N Engl J Med 2007; 356: 348–59. Ullmann AJ, Lipton JH, Vesole DH et al. Posaconazole or fluconazole for prophylaxis in severe graft-versus-host disease. N Engl J Med 2007; 356: 335–47. Murray CJ, Vos T, Lozano R et al. Disability-adjusted life years (DALYs) for 291 diseases and injuries in 21 regions, 1990-2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet 2012; 380: 2197–223. Menzin J, Meyers JL, Friedman M et al. The economic costs to United States hospitals of invasive fungal infections in transplant patients. Am J Infect Control 2011; 39: e15–20. De Pauw B, Walsh TJ, Donnelly JP et al. Revised definitions of invasive fungal disease from the European Organization for Research and Treatment of Cancer/Invasive Fungal Infections Cooperative Group and the National Institute of Allergy and Infectious Diseases Mycoses Study Group (EORTC/MSG) Consensus Group. Clin Infect Dis 2008; 46: 1813–21. Guinea J, Torres-Narbona M, Gijon P et al. Pulmonary aspergillosis in patients with chronic obstructive pulmonary disease: incidence, risk factors, and outcome. Clin Microbiol Infect 2010; 16: 870–7. Patterson JE, Zidouh A, Miniter P, Andriole VT, Patterson TF. Hospital epidemiologic surveillance for invasive aspergillosis: patient demographics and the utility of antigen detection. Infect Control Hosp Epidemiol 1997; 18: 104–8. Garnacho-Montero J, Amaya-Villar R, Ortiz-Leyba C et al. Isolation of Aspergillus spp. from the respiratory tract in critically ill patients: risk factors, clinical presentation and outcome. Crit Care 2005; 9: R191–9. Cunha C, Aversa F, Lacerda JF et al. Genetic PTX3 deficiency and aspergillosis in stem-cell transplantation. N Engl J Med 2014; 370: 421–32. Cunha C, Aversa F, Romani L, Carvalho A. Human genetic susceptibility to invasive aspergillosis. PLoS Pathog 2013; 9: e1003434. von Eiff M, Roos N, Schulten R, Hesse M, Zuhlsdorf M, van de Loo J. Pulmonary aspergillosis: early diagnosis improves survival. Respiration 1995; 62: 341–7. Gerson SL, Talbot GH, Lusk E, Hurwitz S, Strom BL, Cassileth PA. Invasive pulmonary aspergillosis in adult acute leukemia: clinical clues to its diagnosis. J Clin Oncol 1985; 3: 1109–16. Denning DW, Marinus A, Cohen J et al. An EORTC multicentre prospective survey of invasive aspergillosis in haematological patients: Diagnosis and therapeutic outcome. J Infect 1998; 37: 173–80. Greene RE, Schlamm HT, Oestmann JW et al. Imaging findings in acute invasive pulmonary aspergillosis: clinical significance of the halo sign. Clin Infect Dis 2007; 44: 373–9. Cornely OA, Maertens J, Bresnik M et al. Efficacy outcomes in a randomised trial of liposomal amphotericin B based on revised

© 2015 Blackwell Verlag GmbH

Approach to invasive pulmonary aspergillosis

24

25

26

27

28

29 30

31 32

33

34 35

36

37

38

39

40

41

EORTC/MSG 2008 definitions of invasive mould disease. Mycoses 2011; 54: e449–55. Caillot D, Casasnovas O, Bernard A et al. Improved management of invasive pulmonary aspergillosis in neutropenic patients using early thoracic computed tomographic scan and surgery. J Clin Oncol 1997; 15: 139–47. Cornely O, Vehreschild JJ, Vehreschild MJ. http://innere1.uk-koe ln.de/pdf/studien/behandlungspfade/Behandlungspfad_Febrile-Neu tropenie_10.2014.pdf, 2014. Heimann SM, Cornely OA, Vehreschild MJ et al. Treatment cost development of patients undergoing remission induction chemotherapy: a pharmacoeconomic analysis before and after introduction of posaconazole prophylaxis. Mycoses 2014; 57: 90–7. Maertens J, Verhaegen J, Lagrou K, Van Eldere J, Boogaerts M. Screening for circulating galactomannan as a noninvasive diagnostic tool for invasive aspergillosis in prolonged neutropenic patients and stem cell transplantation recipients: a prospective validation. Blood 2001; 97: 1604–10. Ruhnke M, Bohme A, Buchheidt D et al. Diagnosis of invasive fungal infections in hematology and oncology–guidelines from the Infectious Diseases Working Party in Haematology and Oncology of the German Society for Haematology and Oncology (AGIHO). Ann Oncol 2012; 23: 823–33. Schwartz S, Thiel E. Clinical presentation of invasive aspergillosis. Mycoses 1997; 40(Suppl 2): 21–4. Maschmeyer G, Beinert T, Buchheidt D et al. Diagnosis and antimicrobial therapy of pulmonary infiltrates in febrile neutropenic patients–guidelines of the Infectious Diseases Working Party (AGIHO) of the German Society of Hematology and Oncology (DGHO). Ann Hematol 2003; 82(Suppl 2): S118–26. Tacke D, Koehler P, Markiefka B, Cornely OA. Our 2014 approach to mucormycosis. Mycoses 2014; 57: 519–24. Chamilos G, Marom EM, Lewis RE, Lionakis MS, Kontoyiannis DP. Predictors of pulmonary zygomycosis versus invasive pulmonary aspergillosis in patients with cancer. Clin Infect Dis 2005; 41: 60–6. Vehreschild JJ, Ruping MJ, Wisplinghoff H et al. Clinical effectiveness of posaconazole prophylaxis in patients with acute myelogenous leukaemia (AML): a 6 year experience of the Cologne AML cohort. J Antimicrob Chemother 2010; 65: 1466–71. Koehler P, Tacke D, Cornely OA. Our 2014 approach to candidaemia. Mycoses 2014; 57: 581–3. Jang SH, Colangelo PM, Gobburu JV. Exposure-response of posaconazole used for prophylaxis against invasive fungal infections: evaluating the need to adjust doses based on drug concentrations in plasma. Clin Pharmacol Ther 2010; 88: 115–9. Cornely OA, Ullmann AJ. Lack of evidence for exposure-response relationship in the use of posaconazole as prophylaxis against invasive fungal infections. Clin Pharmacol Ther 2011; 89: 351–2. Cornely OA, Helfgott D, Langston A et al. Pharmacokinetics of different dosing strategies of oral posaconazole in patients with compromised gastrointestinal function and who are at high risk for invasive fungal infection. Antimicrob Agents Chemother 2012; 56: 2652–8. Dolton MJ, Ray JE, Chen SC, Ng K, Pont L, McLachlan AJ. Multicenter study of posaconazole therapeutic drug monitoring: exposureresponse relationship and factors affecting concentration. Antimicrob Agents Chemother 2012; 56: 5503–10. Kohl V, Muller C, Cornely OA et al. Factors influencing pharmacokinetics of prophylactic posaconazole in patients undergoing allogeneic stem cell transplantation. Antimicrob Agents Chemother 2010; 54: 207–12. Vehreschild JJ, Muller C, Farowski F et al. Factors influencing the pharmacokinetics of prophylactic posaconazole oral suspension in patients with acute myeloid leukemia or myelodysplastic syndrome. Eur J Clin Pharmacol 2012; 68: 987–95. Duarte RF, Lopez-Jimenez J, Cornely OA et al. Phase 1b study of new posaconazole tablet for prevention of invasive fungal infections in high-risk patients with neutropenia. Antimicrob Agents Chemother 2014; 58: 5758–65.

© 2015 Blackwell Verlag GmbH

42

43

44

45

46 47

48

49

50

51

52

53

54

55

56

57

58

59

60

Walsh TJ, Teppler H, Donowitz GR et al. Caspofungin versus liposomal amphotericin B for empirical antifungal therapy in patients with persistent fever and neutropenia. N Engl J Med 2004; 351: 1391–402. Link H, Bohme A, Cornely OA et al. Antimicrobial therapy of unexplained fever in neutropenic patients–guidelines of the Infectious Diseases Working Party (AGIHO) of the German Society of Hematology and Oncology (DGHO), Study Group Interventional Therapy of Unexplained Fever, Arbeitsgemeinschaft Supportivmassnahmen in der Onkologie (ASO) of the Deutsche Krebsgesellschaft (DKG-German Cancer Society). Ann Hematol 2003; 82(Suppl 2): S105–17. Vehreschild MJ, Vehreschild JJ, Hubel K et al. Diagnosis and management of gastrointestinal complications in adult cancer patients: evidence-based guidelines of the Infectious Diseases Working Party (AGIHO) of the German Society of Hematology and Oncology (DGHO). Ann Oncol 2013; 24: 1189–202. Vehreschild MJ, Meissner AM, Cornely OA et al. Clinically defined chemotherapy-associated bowel syndrome predicts severe complications and death in cancer patients. Haematologica 2011; 96: 1855– 60. Koehler P, Tacke D, Cornely OA. Aspergillosis of bones and joints - a review from 2002 until today. Mycoses 2014; 57: 323–35. Clancy CJ, Jaber RA, Leather HL et al. Bronchoalveolar lavage galactomannan in diagnosis of invasive pulmonary aspergillosis among solidorgan transplant recipients. J Clin Microbiol 2007; 45: 1759–65. Kim SW, Rhee CK, Kang HS et al. Diagnostic value of bronchoscopy in patients with hematologic malignancy and pulmonary infiltrates. Ann Hematol 2015; 94: 153–9. Meersseman W, Lagrou K, Maertens J et al. Galactomannan in bronchoalveolar lavage fluid. Am J Respir Crit Care Med 2008; 177: 27–34. Caillot D, Couaillier JF, Bernard A et al. Increasing volume and changing characteristics of invasive pulmonary aspergillosis on sequential thoracic computed tomography scans in patients with neutropenia. J Clin Oncol 2001; 19: 253–9. Ruping MJ, Vehreschild JJ, Groll A et al. Current issues in the clinical management of invasive aspergillosis–the AGIHO, DMykG, OGMM and PEG web-based survey and expert consensus conference 2009. Mycoses 2011; 54: e557–68. Cornely OA, Arikan-Akdagli S, Dannaoui E et al. ESCMID and ECMM joint clinical guidelines for the diagnosis and management of mucormycosis 2013. Clin Microbiol Infect 2014; 20(Suppl 3): 5–26. Lass-Florl C, Resch G, Nachbaur D et al. The value of computed tomography-guided percutaneous lung biopsy for diagnosis of invasive fungal infection in immunocompromised patients. Clin Infect Dis 2007; 45: e101–4. Walsh TJ, Anaissie EJ, Denning DW et al. Treatment of aspergillosis: clinical practice guidelines of the Infectious Diseases Society of America. Clin Infect Dis 2008; 46: 327–60. Herbrecht R, Denning DW, Patterson TF et al. Voriconazole versus amphotericin B for primary therapy of invasive aspergillosis. N Engl J Med 2002; 347: 408–15. Walsh TJ, Pappas P, Winston DJ et al. Voriconazole compared with liposomal amphotericin B for empirical antifungal therapy in patients with neutropenia and persistent fever. N Engl J Med 2002; 346: 225–34. Cornely OA, Bohme A, Schmitt-Hoffmann A, Ullmann AJ. Safety and pharmacokinetics of isavuconazole as antifungal prophylaxis in acute myeloid leukemia patients with neutropenia: results of a phase 2, dose escalation study. Antimicrob Agents Chemother 2015; Apr;59(4): 2078–85. Ruping MJ, Muller C, Vehreschild JJ et al. Voriconazole serum concentrations in prophylactically treated acute myelogenous leukaemia patients. Mycoses 2011; 54: 230–3. Cornely OA, Maertens J, Bresnik M et al. Liposomal amphotericin B as initial therapy for invasive mold infection: a randomized trial comparing a high-loading dose regimen with standard dosing (AmBiLoad trial). Clin Infect Dis 2007; 44: 1289–97. Cornely OA, Maertens J, Bresnik M, Ullmann AJ, Ebrahimi R, Herbrecht R. Treatment outcome of invasive mould disease after sequential

7

B. Liss et al.

61

62

63

64

65

66

67

68

69

70

71

72

73

74

8

exposure to azoles and liposomal amphotericin B. J Antimicrob Chemother 2010; 65: 114–7. Cornely OA, Vehreschild JJ, Vehreschild MJGT et al. Phase II dose escalation study of caspofungin for invasive aspergillosis. Antimicrob Agents Chemother 2011; 55: 5798–803. Brodoefel H, Vogel M, Hebart H et al. Long-term CT follow-up in 40 non-HIV immunocompromised patients with invasive pulmonary aspergillosis: kinetics of CT morphology and correlation with clinical findings and outcome. AJR Am J Roentgenol 2006; 187: 404–13. Lim C, Seo JB, Park SY et al. Analysis of initial and follow-up CT findings in patients with invasive pulmonary aspergillosis after solid organ transplantation. Clin Radiol 2012; 67: 1179–86. Kim K, Lee MH, Kim J et al. Importance of open lung biopsy in the diagnosis of invasive pulmonary aspergillosis in patients with hematologic malignancies. Am J Hematol 2002; 71: 75–9. Matt P, Bernet F, Habicht J et al. Predicting outcome after lung resection for invasive pulmonary aspergillosis in patients with neutropenia. Chest 2004; 126: 1783–8. Nosari A, Ravini M, Cairoli R et al. Surgical resection of persistent pulmonary fungus nodules and secondary prophylaxis are effective in preventing fungal relapse in patients receiving chemotherapy or bone marrow transplantation for leukemia. Bone Marrow Transplant 2007; 39: 631–5. Mousset S, Buchheidt D, Heinz W et al. Treatment of invasive fungal infections in cancer patients-updated recommendations of the Infectious Diseases Working Party (AGIHO) of the German Society of Hematology and Oncology (DGHO). Ann Hematol 2014; 93: 13–32. Groll AH, Castagnola E, Cesaro S et al. Fourth European Conference on Infections in Leukaemia (ECIL-4): guidelines for diagnosis, prevention, and treatment of invasive fungal diseases in paediatric patients with cancer or allogeneic haemopoietic stem-cell transplantation. Lancet Oncol 2014; 15: e327–40. Spiess B, Seifarth W, Merker N et al. Development of novel PCR assays to detect azole resistance-mediating mutations of the Aspergillus fumigatus cyp51A gene in primary clinical samples from neutropenic patients. Antimicrob Agents Chemother 2012; 56: 3905–10. Hoenigl M, Prattes J, Spiess B et al. Performance of galactomannan, beta-d-glucan, Aspergillus lateral-flow device, conventional culture, and PCR tests with bronchoalveolar lavage fluid for diagnosis of invasive pulmonary aspergillosis. J Clin Microbiol 2014; 52: 2039–45. Willinger B, Lackner M, Lass-Florl C et al. Bronchoalveolar lavage lateral-flow device test for invasive pulmonary aspergillosis in solid organ transplant patients: a semiprospective multicenter study. Transplantation 2014; 98: 898–902. Pazos C, Ponton J, Del Palacio A. Contribution of (1->3)-beta-D-glucan chromogenic assay to diagnosis and therapeutic monitoring of invasive aspergillosis in neutropenic adult patients: a comparison with serial screening for circulating galactomannan. J Clin Microbiol 2005; 43: 299–305. Marty FM, Lowry CM, Lempitski SJ, Kubiak DW, Finkelman MA, Baden LR. Reactivity of (1–>3)-beta-d-glucan assay with commonly used intravenous antimicrobials. Antimicrob Agents Chemother 2006; 50: 3450–3. Pruller F, Wagner J, Raggam RB et al. Automation of serum (1–>3)beta-D-glucan testing allows reliable and rapid discrimination of patients with and without candidemia. Med Mycol 2014; 52: 455–61.

75

76

77

78

79

80

81

82

83

84

85

86

87

88

89

Marchetti O, Lamoth F, Mikulska M, Viscoli C, Verweij P, Bretagne S. ECIL recommendations for the use of biological markers for the diagnosis of invasive fungal diseases in leukemic patients and hematopoietic SCT recipients. Bone Marrow Transplant 2012; 47: 846–54. Hammarstrom H, Kondori N, Friman V, Wenneras C. How to interpret serum levels of beta-glucan for the diagnosis of invasive fungal infections in adult high-risk hematology patients: optimal cut-off levels and confounding factors. Eur J Clin Microbiol Infect Dis 2015. Potenza L, Vallerini D, Barozzi P et al. Mucorales-specific T cells emerge in the course of invasive mucormycosis and may be used as a surrogate diagnostic marker in high-risk patients. Blood 2011; 118: 5416–9. Potenza L, Barozzi P, Vallerini D et al. Diagnosis of invasive aspergillosis by tracking Aspergillus-specific T cells in hematologic patients with pulmonary infiltrates. Leukemia 2007; 21: 578–81. Bacher P, Kniemeyer O, Schonbrunn A et al. Antigen-specific expansion of human regulatory T cells as a major tolerance mechanism against mucosal fungi. Mucosal Immunol 2014; 7: 916–28. Bacher P, Steinbach A, Kniemeyer O et al. Fungus-specific CD4(+) T cells for rapid identification of invasive pulmonary mold infection. Am J Respir Crit Care Med 2015; 191: 348–52. Hamprecht A, Buchheidt D, Vehreschild JJ et al. Azole-resistant invasive aspergillosis in a patient with acute myeloid leukaemia in Germany. Euro Surveill 2012; 17: 20262. Spiess B, Postina P, Reinwald M et al. Incidence of Cyp51 A key mutations in Aspergillus fumigatus-a study on primary clinical samples of immunocompromised patients in the period of 1995-2013. PLoS ONE 2014; 9: e103113. Steinmann J, Hamprecht A, Vehreschild MJ et al. Emergence of azoleresistant invasive aspergillosis in HSCT recipients in Germany. J Antimicrob Chemother 2015. Ananda-Rajah MR, Grigg A, Downey MT et al. Comparative clinical effectiveness of prophylactic voriconazole/posaconazole to fluconazole/itraconazole in patients with acute myeloid leukemia/myelodysplastic syndrome undergoing cytotoxic chemotherapy over a 12-year period. Haematologica 2012; 97: 459–63. Duarte RF, Sanchez-Ortega I, Cuesta I et al. Serum galactomannanbased early detection of invasive aspergillosis in hematology patients receiving effective antimold prophylaxis. Clin Infect Dis 2014; 59: 1696–702. Lerolle N, Raffoux E, Socie G et al. Breakthrough invasive fungal disease in patients receiving posaconazole primary prophylaxis: a 4-year study. Clin Microbiol Infect 2014; 20: O952–9. Winston DJ, Bartoni K, Territo MC, Schiller GJ. Efficacy, safety, and breakthrough infections associated with standard long-term posaconazole antifungal prophylaxis in allogeneic stem cell transplantation recipients. Biol Blood Marrow Transplant 2011; 17: 507–15. Marr KA, Schlamm HT, Herbrecht R et al. Combination antifungal therapy for invasive aspergillosis: a randomized trial. Ann Intern Med 2015; 162: 81–9. Cornely OA, Meems L, Herbrecht R, Viscoli C, vanAmsterdam RG, Ruhnke M. Randomised, multicentre trial of micafungin vs. an institutional standard regimen for salvage treatment of invasive aspergillosis. Mycoses 2015; 58: 58–64.

© 2015 Blackwell Verlag GmbH

Our 2015 approach to invasive pulmonary aspergillosis.

At the University Hospital of Cologne, in general two patient groups at high risk for invasive aspergillosis receive posaconazole prophylaxis: Acute m...
366KB Sizes 1 Downloads 29 Views