<<

PulmonaryMatthias Griese, MD Alveolar Proteinosis: A abstract Comprehensive’ Clinical Perspective Pulmonary alveolar proteinosis is a broad group of rare diseases that are defined by the occupation of a s gas-exchange area by pulmonary that are not properly removed. The clinical and radiologic phenotypes among them are very similar. The age of manifestation plays a central role in the differential diagnosis of the almost 100 conditions and provides an efficient path to the correct diagnosis. The diagnostic approach is tailored to identify genetic or autoimmune causes, exposure to environmental agents, and associations with numerous other diseases. Whole-lung lavages are the cornerstone of treatment, and children in particular depend on the expertise to perform such therapeutic lavages. Other treatment options and long-term survival are related to the condition causing the proteinosis.

3 Under pathological conditions, the inhabitants4 . The first description of alveolar airspaces can be filled with PAP as well as the vast majority of various materials, which frequently data on treatment and prognosis are replace the air necessary for gas related to autoimmune PAP. Most Department of Pediatric Pneumology, Dr von Hauner exchange and give rise to alveolar pediatric cases are nonautoimmune Children’s Hospital, Ludwig-Maximilians-University of Munich and the German Center for Lung Research, Munich, filling syndromes (‍Table 1). These PAP and distribute almost evenly Germany conditions have similar clinical and among many different entities. In radiologic presentations. This makes childhood, there is a bimodal age DOI: https://​doi.​org/​10.​1542/​peds.​2017-​0610 their differential diagnosis difficult. distribution; some conditions manifest Accepted for publication Apr 11, 2017 in the neonatal period (‍Fig 2, upper Address correspondence to Matthias Griese, MD, Pulmonary alveolar proteinosis (‍PAP) portion), whereas others manifest Dr von Hauner Children’s Hospital, University of is defined by the accumulation of during infancy and childhood. Munich, Lindwurmstr 4, 80337 München, Germany. pulmonary surfactants in the alveolar E-mail: [email protected] space (‍Fig 1). Mechanistically, these Pathology PEDIATRICS (ISSN Numbers: Print, 0031-4005; Online, disturbances of homeostasis 1098-4275). Copyright 2017 by the American Academy of may be caused by an altered surfactant1 © production, removal, or both. PAP is The expansion of the surfactant pool Pediatrics can be recognized in histopathological FINANCIAL DISCLOSURE: The author has indicated a heterogeneous group of disorders 2 that is caused by different conditions specimens. The alveoli are filled with he has no financial relationships relevant to this (‍Table 2). The age of manifestation eosinophilic, acellular, and finely article to disclose. plays an important role in the granular material (‍Fig 1E). Clefts FUNDING: Funded by the German Center for Lung Research, DZL 2.0, the European Management diagnostic approach and differential made4,5​ of can be found (‍Fig 1F). ‍ Sometimes, detached type II Platform for Children’s Interstitial Lung Disease diagnosis (‍Fig 2). Success of treatment (FP7, 305653), and E-Rare 2016. and outcome are related to the pneumocytes, foamy macrophages, or granulocytes as well as POTENTIAL CONFLICT OF INTEREST: The author has underlying condition and, in children, to indicated he has no potential conflicts of interest the technical skills needed to perform of normal can be to disclose. whole-lung lavages (‍WLLs). WLL is the identified (‍Fig 1 C and D). cornerstone of PAP treatment. Epidemiology Whereas the intra-alveolar filling does To cite: Griese M. Pulmonary Alveolar Proteinosis: not differentiate between different A Comprehensive Clinical Perspective. Pediatrics. forms of PAP, histopathological 2017;140(2):e20170610

PAP is a rare disease complex examination of the alveolar wall that affects <5 cases per 100000 structures may be normal (‍Fig 1E) Downloaded from www.aappublications.org/news by guest on October 1, 2021 PEDIATRICS Volume 140, number 2, August 2017:e20170610 STATE-OF-THE-ART REVIEW ARTICLE or abnormal. The interstitial space idiopathic occurs in children, and it needs is mostly normal; however, several in a mature neonate. to be differentiated from GM-CSF forms show4, 6​widening from cells Investigations and Differential antibodies in healthy individuals or fibrosis. ‍ When the surfactant Diagnosis at low levels and patients with “ dysfunction syndromes were first autoinflammatory diseases such as ” Chest Radiography – 21 described, the term congenital colitis, Crohn disease, malignancies,22 24​ alveolar proteinosis7 was used or PAP caused by dust exposure. ‍ in such newborns. Particularly Because there is a close correlation in children, the PAP histopattern The presence of diffuse bilateral between antibody level and PAP mostly symmetrical alveolar, 25,26​ presents in combination 8,with9​ development,​ ‍ a close link additional histopatterns,​ ‍ such sometimes patchy infiltrates with to pulmonary manifestation is as increased cellularity from air bronchograms give first clues mandatory when searching for these to interstitial lung disease due to hyperplasia of type II pneumocytes autoantibodies.Lactate Dehydrogenase and collagen fibers or alveolar alveolar filling (‍Fig 1A). Usually, ∼ 10 macrophages. In cohort studies in the perimediastinal regions are 50% to 60% of such biopsies, the more affected than the subpleural 11,12​ Lactate dehydrogenase is increased PAP histopattern was found,​ ‍ regions. Persistence of the cloudy in 82% of patients with autoimmune and the detection rate was higher in infiltrates after antibiotic treatment 6 is a frequent observation. In neonates PAP. Of interest, this marker biopsies conducted during the early “ ” 12 readily responds to changes in disease phase. In TTF1 deficiency, with acute RDS, all radiologic stages – disease severity and improves after pulmonary histology may also show (‍including a white lung ) may 6,27​ 29 develop progressively. therapeutic lavages. ‍ ‍ the PAP pattern in addition to the Lung Function Tests nonspecific interstitial Similarly, in serum carcinoembryonic antigen, levels of the surfactant pattern and defects13 in lung 30 A, D,​ and KL-6 are markers development. Adults and children who are old of disease activity, which can be Clinical Presentation enough to perform have used to monitor its course. Serum a restrictive pattern with small KL-6 accurately predicts disease and reduced diffusing 29 – progression in autoimmune PAP. PAP presents in 2 types: Most capacity of the lung for carbon frequently (‍70% 90% of PAP cases), monoxide, although 6initial tests In children and adults, autoantibodies patients develop slowly increasing may also be normal. Of interest, including antinuclear antibody, dyspnea (‍initially2 on exertion) and carbon dioxide elimination rarely is antineutrophil cytoplasmic antibody – dry coughing. In PAP caused by a problem in the face of significant (‍eg, cANCA/PR3 and pANCA/ granulocyte-macrophage colony- hypoxemia. At presentation, 55% MPO), anti double-stranded DNA, stimulating factor receptor alpha of the patients with PAP caused anticentromere, cyclic citrullinated – – (‍GM-CSF-Ra) mutations, 70% had by GM-CSF-Ra mutations had peptide, extractable nuclear antigen 14 – dyspnea at presentation, 15% had hypoxemia,​ whereas in adults with (‍eg, anti SS-A [Ro], anti SS-B [La], tachypnea, 30% had clubbing, 35% autoimmune PAP, hypoxemia at anti-RNP, anti Jo-1, anti-Sm, and Scl- had global , and 14 rest was present in approximately 70), and rheumatoid factor should be 15% were intubated and ventilated. – one-third and during exercise 3in assessed. more than half of the patients. With Global and specific immunologic Less frequently (‍30% 50% of PAP treatment of the alveolar filling, function tests need to be done cases), , weight loss, fatigue, abnormal lung function tests are to diagnose underlying immune and chest pain are observed expected to be reversible; if they are Geneticdeficiencies Testing (‍Table 2). (‍numbers2 are for autoimmune not, then additional pathology should PAP ). Among the children, 45% had beLaboratory suspected. Blood Tests infections (‍mycoplasma, , or respiratory syncytial virus) Autoantibodies Against GM-CSF In neonates with the characteristic before PAP because of GM-CSF-Ra clinical and radiologic presentation mutations, 26% had a , 5% (‍and after the exclusion of other, had a fever,14 and 36% had failure In all cases of suspected PAP, these– more frequent causes), testing for to thrive. PAP caused by affected should be searched. In adults, >90%15 19 mutations in SFTPB, SFTPC, ABCA3, surfactant production typically of the case results are positive. ‍ and TTF1 is recommended (‍www.​ presents with idiopathic respiratory Serology has excellent20 sensitivity childeu.​net). In infants and older distress syndrome (‍RDS) or and specificity. Autoimmune PAP patients, analysis for mutations in Downloaded from www.aappublications.org/news by guest on October 1, 2021 2 Griese TABLE 1 Intra-Alveolar Filling With Different Materials Leads to Various Disease Entities Material Filling the Alveolar Space Disease excluding cardiac, infectious, central, Surfactant PAP or metabolic causes). In older infants Erythrocytes, siderophages Alveolar hemorrhage Macrophages Desquamative interstitial pneumonia and children, red flags include slowly Hyaline membranes , RDS developing dyspnea or dyspnea Eosinophils persisting after resolution of an acute Cholesterol crystals, foreign material Exogenous pneumonia, infection, together Exsudative neutrophilic alveolitis, progressively Meconium aspiration syndrome with diffuse alveolar infiltrates on the replaced by macrophages, giant cells, fibroblasts , Pneumocystis P jiroveci pneumonia chest radiograph. Calcified microliths Pulmonary alveolar microlithiasis Blood tests are done first, followed Plasma, serum Pulmonary by a bronchoscopy with standardized lavage and transbronchial biopsy in most patients (‍Table 2). When the – results obtained are not definitively SFTPC, ABCA3, CSF2RB, CSF2RA, , , lipoid diagnostic, a lung biopsy is needed to GATA2, SLC7A7, methionyl transfer pneumonia, and Niemann-Pick31 firmly establish the diagnosis. RNA synthetase (‍MARS), NPC2, and disease, among others. Etiology possibly NPB (‍see Table 2) should be It must be kept in mind that done when appropriate. Bronchoalveolar Lavage computed tomography (‍CT) imaging in neonates and infants with high Categorization of PAP forms according rates is very demanding to suspected etiology is helpful With standardized diagnostics, ü and needs a lot of technical detail to for therapy, although frequently lavage return typically gets milkier obtain high-quality scans. Therefore, the underlying etiology cannot be on visual inspection. May-Gr nwald CT scanning should be reserved for Surfactantidentified precisely Dysfunction (‍Table Syndromes 2). Giemsa staining shows cellular the specialized center performing debris, acellular globules, and foamy diagnostic imaging in temporal macrophages (‍Fig 1 C and D). Lungrelation Biopsy to the lung biopsy. Mutations in SFTPB, SFTPC, In adults with PAP, experienced ABCA3, and TTF1 may present with centers can make the diagnosis by – the histologic PAP pattern mostly bronchoalveolar lavage (‍BAL) in 7,9,​ 32​ 34 In infants, biopsies are done during the neonatal period,​ ‍ ‍ ‍ 70% to 74% of the patients with 3,16​ as open biopsies or sometimes when secretion of surfactant is acquired PAP. ‍ BAL fluid should be thoracoscopically. In adults and older normally increased more than investigated for infections (‍Table 2). 35,36​ High-Resolution Computed children, transbronchial biopsies can 10-fold ‍ and sometimes later as ∼ 37,38​ Tomography be done during a bronchoscopy. Lung well but to a smaller degree. ‍ biopsies were performed in 80% At the same time, the removal of

of the children14 to make the diagnosis aberrant surfactant may be impaired Crazy paving pattern, which of PAP. In children, reference because of volume- and39 barotrauma- combines ground-glass opacity (‍a reading of the biopsy specimens by induced lung injuries,​ and hazy increase in lung opacity that a specialized pathologist is highly physiologically increased removal does not obscure the underlying recommended and can be organized may be disturbed, all of which lead via different networks, such as the to swamped macrophage-removal vessels or bronchial structures) and ’ 40 a superimposed reticular pattern (‍Fig European Management Platform for systems. If patients survive, the 1B), is characteristic for PAP but not Children s Interstitial Lung Diseases PAP pattern slowly disappears and is specific to it. Its differential diagnosis (‍www.​childeu.​net). replaced by a more10,12​ fibrotic pattern includes edema, hemorrhage, acute Diagnosis of PAP of lung disease. ‍ Similarly, such a RDS, acute interstitial pneumonia, sequence has been demonstrated41 for MARS mutations (‍Table 2). Today, eosinophilicPneumocystis pneumonia, jirovecii diffuse alveolar damage, As with all rare lung diseases, a high fortunately, conditions are diagnosed caused by , degree of suspicion is necessary genetically, which eliminates the cytomegalovirus, adenovirus, when taking the diagnosis of PAP into need for a biopsy. mycoplasma, bacteria, tuberculosis, consideration. Red flags in neonates Impaired GM-CSF Signaling nonspecific interstitial pneumonitis, and young infants are nonimproving organizing pneumonia, eosinophilic or slowly improving respiratory granulomatosis with polyangiitis, distress in the mature neonate (‍ie, if GM-CSF is critical for the regulation radiation pneumonitis, drug-related persistent after 1 or more weeks and of surfactant homeostasis, alveolar Downloaded from www.aappublications.org/news by guest on October 1, 2021 PEDIATRICS Volume 140, number 2, August 2017 3 FIGURE 1 Diagnosing alveolar pulmonary proteinosis. A, A chest radiograph shows bilateral, often symmetrical alveolar opacities. If a radiograph appears less dense than what is shown here, a “butterfly” appearance may be recognized. B, A crazy paving pattern from ground-glass opacification and overlaid reticulonodular pattern is shown. C, Foamy macrophage from BAL filled with lipid material (May-Grünwad staining) is seen. D, Periodic acid–Schiff (PAS) staining–positive noncellular globules. A large amount of cell debris is characteristic of PAP lavage. E, Histology from a patient with PAP caused by a homozygous GM-CSF-Ra mutation is shown. Note the alveolar filling and normal alveolar walls (haematoxylin-eosin staining). F, The same biopsy with a PAS stain shows amorphous PAS-positive material with abundant oval bodies.

43 “ macrophage maturation and GM-CSF signaling by neutralizing PAP. This autoimmune disease, in ” “ ” phagocytosis, lung host42 defense, GM-CSF autoantibodies is the cause the beginning called adult idiopathic and innate immunity. DisruptedDownloaded fromof www.aappublications.org/newsthe vast majority of adult bycases guest of on OctoberPAP 1, 2021 or primary PAP,​ is now called 4 Griese TABLE 2 Categorization of PAP Disease Causes of PAP Manifestation Genetically Exposure Histologically Intact Reported in Causedb Caused Lung Interstitial Tissue, Neonates, Primarily Alveolar Filling Childrena Surfactant-dysfunction syndromes SFTPB mutations Y Y N N SFTPC mutations Y Y N N ABCA3 mutations Y Y N N TTF1 mutations Y Y N N Impaired GM-CSF signaling GM-CSF receptor α chain of mutations Y Y N Y Turner syndrome with heterozygous GM-CSF receptor α chain mutations Y Y N Y GM-CSF receptor β chain mutations Y Y N Y Autoimmune GM-CSF antibodies Y N N Y Hematologic disorders and other malignancies GATA2 deficiency Y Y N Y MDS (most common) Y Y N Y Chronic myelomonocytic leukemia Y Y N n.k. Acute lymphatic leukemia Y Y N Yc Congenital dyserythropoietic anemia Y Y N Y Fanconi’s anemia Y Y N Y Hemophagocytic lymphohistiocytosis Y N N Sideroblastic anemia Y Nc N Yc Primary myelofibrosis, chronic lymphocytic leukemia, cutaneous -cellT N Y or n.k. Nc Yc or n.k. lymphoma, thymic alymphoplasia, adult T-cell leukemia and/or lymphoma, idiopathic thrombocytopenic purpura, aplastic anemia, chronic myeloid leukemia, overlap myeloproliferative neoplasm, acute myeloid leukemia, hairy-cell leukemia, multiple myeloma and/ or plasmocytoma, polycythemia vera, essential thrombocythemia, amyloidosis, Hodgkin disease, non-Hodgkin lymphoma, adenocarcinoma, glioblastoma, melanoma, small-cell lung carcinoma, clear-cell renal cell carcinoma, Systemic diseases Lysinuric intolerance (SLC7A7 mutation) Y Y N Y (50%), N (50%) MARS mutations Y Y N N Niemann Pick type C2 Y Y N N Niemann Pick type B Y Y N n.d. Bone marrow, stem cell transplant Y N N Yc Systemic lupus erythematosus, granulomatosis with polyangiitis, N N N Yc or N microscopic polyangiitis, membranous nephropathy, dermatomyositis with interstitial lung disease, coincident in many other rheumatologic diseases and interstitial lung diseases, lung transplant Immunologic diseases Adenosine deaminase deficiency Y Y N Y Agammaglobulinemia Y Yc N n.k. DiGeorge syndrome type 2 Y Y N n.d. Monoclonal gammopathy N N N Y Selective immunoglobulin A deficiency N N N Y Severe combined immunodeficiency Y Y N Y X-linked hyper–immunoglobulin M syndrome Y Y N n.d. Infections Cytomegalovirus Y N Y N or Nc Epstein-Barr virus Y N Y N or Nc HIV Y N Y N or Nc M tuberculosis Y N Y N or Nc Atypical mycobacteria Y N Y N or Nc Nocardia N N Y N or Nc P jiroveci Y N Y N or Nc Drugs Chemotherapy, antineoplastic Y N Y Y

Downloaded from www.aappublications.org/news by guest on October 1, 2021 PEDIATRICS Volume 140, number 2, August 2017 5 TABLE 2 Continued Disease Causes of PAP Manifestation Genetically Exposure Histologically Intact Reported in Causedb Caused Lung Interstitial Tissue, Neonates, Primarily Alveolar Filling Childrena Busulfan, sirolimus, everolimus, tyrosine kinase inhibitors (including N N Y Y imatinib, nilotinib, and dasatinib), mycophenolate and cyclosporine combination, smoked fentanyl patches, leflunomide, hydrofluoric acid (inhaled) Types of dust exposure, inorganic Aluminum, cement, marble, indium, iron, silica and silica-leaking breast N N Y N (except aluminum, implants, tin, titanium (and varnish) silica) Types of dust exposure, organic Bakery flour, chlorine, cleaning products, cotton, fertilizer, agricultural N N Y N or Nc dust, fumes, gasoline, hydrofluoric acid, parrots, pigeons, petroleum, sawdust Miscellaneous conditions Osteopetrosis caused by TCIRG1 gene mutation Y Y N Yc Total anomalous pulmonary venous return with coarctation of the aorta Y N, Yc N Y, pulmonary hypoplasia (single case) Y or N indicates highest likelihood and/or firm knowledge from publications; frequently extensive data are lacking and these indications are based on estimation. n.d., not done; n.k., not known. a Manifestation of PAP is proven in this age group (not of disease). b May be germline or clonal in monocytes. c Indicates uncertainty.

“ ” 44 autoimmune PAP ‍ and can also occur in children. Interruption of GM-CSF signaling by α β hereditary factors, such as mutations in the (‍CD116) or (‍CD131) chain of the GM-CSF receptor (‍which is also used by the interleukin-345,46​ and interleukin-5 receptors),​ ‍ will all lead to hereditary PAP forms. These cases most likely manifest in childhood14, but47,​ 48​ may also be diagnosed inHematologic adults. ‍ Disorders and Other Malignancies

Hematologic disorders widely contribute 49to PAP causes,50 both in children and adults (‍Table 2). Prolonged neutropenia and reduction of alveolar macrophages FIGURE 2 by myeloablative chemotherapy Schematic of age dependency of the manifestation of various alveolar pulmonary proteinosis forms or leukemic infiltration have been and qualitative estimates of age at clinical manifestation in the major groups of PAP. Note the size of implicated in the pathogenesis of the graphs does not represent their absolute frequency. Autoimmune PAP represents ∼90% of all 51 cases if age of manifestation is not considered. pediatric PAP. Autoantibodies against GM-CSF may be elevated in BAL fluid, whereas in sera, 52 – they were below sensitivity. ∼ – Among patients with GATA2 myeloid leukemia (‍14%); papillomavirus or Epstein- haploinsufficiency, 20% chronic myelomonocytic Barr virus associated tumors; develop PAP, often in association leukemia (‍8%); severe infections venous thrombosis; lymphedema; with familial myelodysplastic (‍including mycobacterial and sensorineural hearing loss; 53 syndrome (‍MDS) (‍84%); acute fungal infections); human miscarriage; and hypothyroidism. Downloaded from www.aappublications.org/news by guest on October 1, 2021 6 Griese Systemic Diseases

opportunistic infections between one of the lungs is ventilated and 1950 and 2010 were searched, and the other is filled with aliquots of Lysinuric protein intolerance is 75 cases were reviewed. Forty-three warmed saline by infusion with an inherited defect of cationic percent of the patients had nocardia gravity or gentle pressure via a amino acid (‍lysine, arginine, Mycobacteriuminfection, 37% had tuberculosis mycobacterial 50-mL syringe. After recovery of the15 and ornithine) transport at the infection (‍of those, 75% were fluid, the next washing cycle begins. basolateral membrane of epithelial ), 61 For smaller children or infants, we and 20% had fungal infections. have developed and extensively cells and is54 caused by mutations 65 in SLC7A7. The condition can Opportunistic infections can either tested a novel technique. In this – be considered a disorder of bone precede or follow a diagnosis of technique, 1 lung is occluded with a marrow derived monocytes PAP. PAP should be considered pulmonary artery balloon catheter in apparently immunocompetent (‍which is continuously watched with differentiating into dysfunctional55 alveolar macrophages. Lung patients who present withPneumocystis an a small endoscope for its tight fit into involvement was observed in 71% of opportunistic infection and diffuse the ) and used for lavage children, which showed fibrosis on alveolar infiltrates. For of that lung,27, and66​ the other lung is and the viruses indicated in Table ventilated. ‍ Indication for WLL is histopathology56 or CT scan in addition to PAP. 2, individual cases of PAP were given when respiratory symptoms reported. impair the quality of life (‍eg, oxygen Drugs and Dust Exposure MARS deficiency by missense treatment is necessary), development é mutations leads to PAP, which is of weight is impaired, or lung endemic on R union Island in the functionExtracorporeal deviates M fromembrane normal. 41 Oxygenation and Lung Transplants Indian Ocean. The disease starts Although PAP caused by dust in infancy with PAP then frequently exposure has not been reported in children, a detailed occupational progresses to pulmonary fibrosis57 with cholesterol granulomas. and environmental history has to Sometimes, severe hypoxemia be taken for diagnoses in every precludes immediate WLL, and new patient regardless of age. In patients need to be supported Niemann-Pick diseases are adults, between 23% and 39% of by extracorporeal membrane hereditary neurovisceral lysosomal patients with PAP were exposed to oxygenation to allow for the lipid-storage disorders, which can 2,3​ compounds shown to induce PAP. ‍ procedure and recovery or to have present with respiratory symptoms Miscellaneous Conditions it serve as a bridge to the lung at any age. In type C2, PAP may be 65 transplant. Several patients caused by reduced NPC2 protein with PAP (‍in its various forms) expression in alveolar macrophages. have had a lung transplant. The composition and function of Any disease involving the lungs However, the recurrence of PAP surfactants in this form of PAP are may potentially cause PAP, and thus 58 disease has been reported in some abnormal. PAP has been reported PAP needs to be included in the 67,68​ patients. ‍ in Niemann-Pick type B; however, it differential diagnosis. Alternatively, Natural Course, Specific was unclear if this was a coincidence PAP could be coincident with another 59 Treatment Options, and Outcome Immunologicwith autoimmune Diseases PAP. lung disease; thus diagnostic workup62 for different causes is mandatory. General Treatment Approach Each etiologic group of PAP has its ∼ Of infants with adenosine deaminase Therapeutic WLL own clinical course, treatments and deficiency, 50% have PAP with outcome, and due to rarity of cases, typical lung pathology and without has not been described in detail for additional abnormalities, except for WLL remains the current standard most.Autoimmune PAP coincident 60identification of various of care for PAP. Over the years, the pathogens. For several other technique has been6, 63,​improved64​ in immunologic diseases, cases of PAP dedicated centers. ‍ ‍ Bilateral In autoimmune PAP, historical comparisons suggest that patients Infectionshave been reported (‍Table 2). sequential WLL in the same treatment session is aimed at, but is before the advent of therapeutic dependent on, the clinical condition lung lavages had higher mortality than after therapeutic lung lavages of the patient. In adults and older ∼ In a recent review of the literature, children in whom a double-lumen had become6 an established all reported cases of PAP and tube can be safely placed repetitively, treatment. In 5% to 10% of Downloaded from www.aappublications.org/news by guest on October 1, 2021 PEDIATRICS Volume 140, number 2, August 2017 7 PAP Caused by Hematologic ∼ Disorders patients, spontaneous3,6​ remission necessary until the effluent is clear may occur,​ ‍ and in 50%, only 1 (‍Fig 3 D and E). The outcome depends ∼ WLL treatment is necessary for long- mainly on WLL treatment, which 2 These patients present with term remission. Mortality during 16 was successfully performed14 in 90% follow-up in large centers is <10%. of all published cases. In these respiratory insufficiency and fever in WLL is the standard treatment; forms of PAP, hematopoietic stem 24% of cases, particularly in patients with prolonged85 neutropenia from however, additional69 options have cell transplants may be successful to been developed. To overcome the correct a defect that is localized in chemotherapy,​ which demands endogenous GM-CSF autoantibodies alveolar macrophages. A recurrence suspicion for this complication. Initial attempts to treat PAP caused by MDS that neutralize GM-CSF, recombinant of PAP in such a child (‍M. Castelle, 52 with WLLs were not successful,​ and exogenous GM-CSF has been MD, personal communication, 2015) 50 69,70​ although later survival improved. tested. ‍ Administration of or an inadvertent45 outcome have been aerosolized GM-CSF appeared more reported ; however, this scenario WLL may be used as a bridge to bone marrow transplants, as reported for attractive and more effective in a needs to be investigated further. 86 controlled prospective trial of 50 an adult patient and also recently PAP Caused by Affected Surfactant for a small 6-year-old child, for whom patients71,72​ with a response rate of 62%. Of interest are studies with Production we performed 14 WLLs over a period a goal of decreasing the GM-CSF of 1 year until a suitable donor was found. PAP resolved rapidly after the amounts necessary by combining 49 73 transplant. The amount of protein it with WLL. Another approach Several infants previously labeled recovered in this condition was combined WLL and plasmapheresis as having congenital PAP have been similar to the amount in autoimmune to reduce the circulating level of treated with limited success or 6 PAP or GM-CSFR mutations (‍Fig 3). GM-CSF autoantibodies; however, without success by WLLs. An infant the results for clinical74,75​ efficacy with PAP likely caused by a surfactant- 87 – – Xue et al described a patient with are controversial. ‍ Rituximab, dysfunction disorder was82 treated a humanized B-lymphocyte with liquid ventilation. An infant girl PAP and MDS due to idic(‍20q ) who depleting antibody, is able to reduce with familial congenital PAP who was was too sick to be lavaged. Consider a neutralizing GM-CSF autoantibodies– not responsive to lavages recovered timely transfer of patients to centers experienced in WLL technique. and showed improved lung 2,function76​ 78 3 times from respiratory failure in high-resolution CT scans. ‍‍ after intravenous immunoglobulin G An important report indicates that There is no rationale for all the latter administration;83 a mutation in SFTPB approaches in nonautoimmune PAP. was excluded. PAP reports from empirical treatment GM-CSF Receptor α or β Chain in PAP secondary to MDS is a risk Mutation–Caused PAP children with molecularly defined surfactant-dysfunction disorders factor for infection88 and contributes are scarce. In our cohort of patients to poor prognosis. We had a similar with SFTPC mutations, we reported experience in a patient with PAP 27 WLL treatment is the mainstay of caused by a GM-CSF-Ra mutation,​ therapy in patients with receptor 5 patients suffering from PAP 12who have been treated with WLLs. No which suggests the need for caution mutations and has been used in the with immunosuppressive therapy in majority of cases reported so far clinical success was observed81 in a 14 these conditions. In an adult patient (‍78%). In some patients with the child with mutation C121F,​ whereas Mycobacterium reasonable success was observed aviumwith PAP intracellulare caused by GATA2 deficiency same mutation, treatment every 4 and infection with in a patient with8 severe course and to 8 weeks27 over several years was mutation I73T. The amount of 49 , WLL was not necessary,​ whereas in others, only helpful. few or no lavages were sufficient in protein recovered in these conditions maintaining a stable clinical course. was much lower than in other PAPs (‍Fig 3). This clearly indicates that The outcome of these conditions The protein amounts recovered from in addition to alveolar filling with is strongly determined by the the lungs were similar to those79 in surfactants, other disease mechanisms underlying condition and is generally adults with autoimmune PAP (‍Fig 3). play a major84 role for respiratory poorer than in autoimmune PAP. WLLs within a few days are less failure. We are not aware of WLLs Median survival time was only50,88​ 20 efficient than lavages repeated at months in a Japanese cohort ‍ ; in in patients with mutations in SFTPB, ∼ least 2 or more weeks apart (‍Fig 3C). ABCA3, or TTF1. The outcomes of another cohort, the death16 rate in such The amount of protein recovered these patients are only initially related subjects was 50%. The causes over time changes with disease to PAP severity; and later, pulmonary of death were hematologic disease activity and with the lavage volume fibrosis predominates. (‍33%), respiratory insufficiency Downloaded from www.aappublications.org/news by guest on October 1, 2021 8 Griese FIGURE 3 Analytical results of WLL effluents in patients with different molecularly defined forms of PAP. A, The total amount of protein removed from the left lung (LL) and right lung (RL) during repetitive WLL. Each dot indicates a single WLL. B, Washout kinetics of protein concentration in lavages from different patients are shown. GM-CSF-Ra #1, n = 26; GM-CSF-Ra #2, n = 28; MDS DiGeorge, n = 9; NPC2, n = 4; SP-C, n = 11. C, WLL protein removed varies with intervals used until next lavage. From day 0 through 180, lavage intervals were 1 to 4 days, then 4 weeks, then 1 to 4 days, etc. Note the low amount of protein removed after a short interval from the previous lavage. From day 180 through 540, lavage intervals were always at least 2 weeks and usually 3 to 4 weeks. D, Over- time variation of volume necessary until clear effluent is shown. E, Over-time variation of protein washed out is shown. The detailed descriptions of the PAP cases used here can be found for GM-CSF-Ra -1 in Griese et al,​27 for GM-CSF-Ra -2 in Hildebrandt et al14 (subject I), for NPC2 in Reunert et al80 (subject 1), for SP-C (I73T) in Brasch et al,8​ for SP-C (C121F) in van Hoorn et al,81​ and for MDS DiGeorge2 in Griese et al49 (subject 7). Ethics approval is documented in the individual studies.

41 (‍25%), infection (‍25%), and WLL has been described in a PAP PAP. Patients develop PAP early unspecified bleeding (‍13%). associated with lysinuric protein in their disease course, and several Systemic Diseases 55 57 intolerance. This suggests that in received empirical WLLs. An Lysinuric Protein Intolerance (SLC7A7 selected cases, WLL may be helpful and analysis of the cohort of 34 patients Mutation) could be attempted by experienced suggested that WLL did not57 have an specialists. GM-CSF89, therapy90​ may be an influence on survival rates. The additional option. ‍ protein content, the composition, MARS Mutations Children present with failure to thrive and the concentration of surfactant and gastrointestinal symptoms from proteins A, B, C, and D as well as lipid pancreatic insufficiency. In 2 children composition and material removed

<2 years old, WLLs56 were performed Recently, we identified biallelic from the lungs were not different from without efficacy. However, at least missense mutations in MARS as the autoimmune PAP (‍M.G., unpublished 1 case of long-lasting remission after cause of a specific form of pediatric observations). Overall outcome of this Downloaded from www.aappublications.org/news by guest on October 1, 2021 PEDIATRICS Volume 140, number 2, August 2017 9 Drugs ö ∼ condition is poor, with57 a median age at K. Reiter, C. Sch n, F. Hoffmann, A. death of 17 years. Schams, T. Wesselak, E. Kaltenborn, Niemann Pick Type C2 After 9 months of treatment with M. Kappler, J. Ripper, J. Hildebrandt, sirolimus, PAP was diagnosed in P. Lohse, F. Brasch, M. Hofmann, I. a heart-lung transplant patient. Eckerlein, and the many coworkers The drug was discontinued, Early lung disease in patients with on published research projects and GM-CSF therapy was started, Niemann-Pick disease type C2 may the cooperators in internal medicine 58,80​ and 3 WLLs were performed to be associated with PAP. ‍ In Griese 94 and including M. 58 80 relieve respiratory distress. A et al and Reunert et al,​ empirical – Luisetti, I. Campo, S.A. Papiris, E.D. patient treated with the disease- treatment with WLL in those patients Manali, U. Costabel, and F. Bonella. modifying anti rheumatoid arthritis was only transiently effective, Finally, I thank the families and drug leflunomide developed PAP reducing the need for oxygen and children for their longstanding (‍verified by a biopsy specimen) improving imaging. The amount of confidence. I apologize to other and was treated with WLL and protein recovered was low, which researchers of many excellent articles subsequently discontinued taking the also differentiated this form of PAP 95 and reviews of the condition who leflunomideDust Exposure. from the others (‍Fig 3 A and B). were not included because of limited Such treatments may be helpful in space. less severe cases or when bridging Abbreviations WLL may be symptomatically to other treatments is the goal. The beneficial, but stopping exposure is outcome is frequently determined by pivotal. WLL may be ineffective after other organ complications from the BAL: bronchoalveolar lavage long-standing or extensive exposure, underlying condition. CT: computed tomography Immunologic Diseases as additional pathologies such as GM-CSF: granulocyte-macro - cholesterol granuloma and fibrosis 96 phage colony-stimulat- may have developed. – ing factor Conclusions PAP caused by immunologic MARS: methionyl transfer RNA diseases is best treated by synthetase correcting60, the91​ underlying defect MDS: myelodysplastic syndrome if possible ‍ or by symptomatic The label PAP is merely an overall PAP: pulmonary alveolar treatment (‍eg, immunoglobulin description of alveolar filling with proteinosis therapy). A 15-year-old boy with surfactant. It is very important RDS: respiratory distress agammaglobulinemia and recurrent to classify all cases definitively syndrome according to etiology and on the PAP was treated with additional 92 WLL: whole-lung lavage basis of new knowledge generated WLL without any complications. References A 6-year-old child we treated with by translational research. PAP of all bridging WLL until a successful etiologies known may be observed 1. Griese M. Pulmonary surfactant in health and human lung diseases: stem cell transplant was performed in children. The diagnostic algorithm state of the art. Eur Respir J. had underlying syndromic as well as treatment options strongly depend on age at presentation (‍Fig 2, 1999;13(6):1455–1476 immunodeficiency associated49 with DiGeorgeInfection syndrome type 2. ‍Table 2). 2. Bonella F, Theegarten T, Guzman J, Acknowledgments Costabel U. Alveolar lipoproteinosis “ syndromes. Eur Respir Mon. ” 2011;54:171 186 There is an ongoing chicken or the – egg debate as to whether specific This review is based on my 3. Inoue Y, Trapnell BC, Tazawa R, infections in PAP are causes or experience as a pediatric et al; Japanese Center of the Rare Lung Diseases Consortium. symptoms, which may be clarified by pulmonologist caring for children of Characteristics of a large cohort therapeutic intervention. The case of all ages with PAP, my experience as a of patients with autoimmune a 3-year-old boy with histologically researcher interested in PAP running pulmonary alveolar proteinosis in proven PAP, a high load of Epstein- a laboratory for the determination of Japan. Am J Respir Crit Care Med. Barr virus, and a response to GM-CSF autoantibodies, and a review 2008;177(7):752–762 ganciclovir treatment supports the of the literature on PAP. Diagnosis 4. Rosen SH, Castleman B, Liebow AA. view that specific93 infections may and treatment of PAP are the result Pulmonary alveolar proteinosis. N Engl induce PAP. Early WLL may reduce of a team approach and would not J Med. 1958;258(23):1123–1142 complications by infections from have been possible without the many nocardiosis or mucormycosis. people involved, including T. Nicolai, Downloaded from www.aappublications.org/news by guest on October 1, 2021 10 Griese 5. T ravis WD, Colby TV, Koss MN, Rosado- a single-center cohort of 70 patients. congenital alveolar proteinosis. BMC de-Christenson ML, Müller NL, King TE. Respir Med. 2011;105(12):1908–1916 Pediatr. 2011;11:72 Non-Neoplastic Disorders of the Lower 17. Manali ED, Papadaki G, Konstantonis D, 28. Seymour JF, Doyle IR, Nakata K, et al. Respiratory Tract. Washington, DC: et al. Cardiovascular risk in pulmonary Relationship of anti-GM-CSF antibody American Registry of Pathology; 2002 alveolar proteinosis. Expert Rev Respir concentration, surfactant protein 6. Seymour JF, Presneill JJ. Pulmonary Med. 2016;10(2):235–240 A and B levels, and serum LDH to alveolar proteinosis: progress in the 18. Papiris SA, Tsirigotis P, Kolilekas L, pulmonary parameters and response first 44 years. Am J Respir Crit Care et al. Pulmonary alveolar proteinosis: to GM-CSF therapy in patients with Med. 2002;166(2):215–235 time to shift? Expert Rev Respir Med. idiopathic alveolar proteinosis. Thorax. 2003;58(3):252 257 7. Nogee LM, de Mello DE, Dehner LP, 2015;9(3):337–349 – Colten HR. Brief report: deficiency of 29. Bonella F, Ohshimo S, Miaotian C, 19. Latzin P, Tredano M, Wüst Y, et al. pulmonary in Anti-GM-CSF antibodies in paediatric Griese M, Guzman J, Costabel U. congenital alveolar proteinosis. N Engl pulmonary alveolar proteinosis. Serum KL-6 is a predictor of outcome J Med. 1993;328(6):406 410 in pulmonary alveolar proteinosis. – Thorax. 2005;60(1):39–44 Orphanet J Rare Dis. 2013;8:53 8. Brasch F, Griese M, Tredano M, et al. 20. Uchida K, Nakata K, Carey B, et Interstitial lung disease in a baby with al. Standardized serum GM-CSF 30. Griese M, Lorenz E, Hengst M, a de novo mutation in the SFTPC gene. autoantibody testing for the et al. Surfactant proteins in pediatric Eur Respir J. 2004;24(1):30–39 routine clinical diagnosis of interstitial lung disease. Pediatr Res. 2016;79(1 1):34 41 9. Brasch F, Schimanski S, Mühlfeld C, autoimmune pulmonary alveolar – – et al. Alteration of the pulmonary proteinosis. J Immunol Methods. 31. Webb WR, Müller NL, Naidich DP. surfactant system in full-term 2014;402(1–2):57–70 High Resolution CT of the Lung. infants with hereditary ABCA3 21. Ser geeva A, Ono Y, Rios R, Molldrem 4th ed. Netherlands: Wolter Kluver; deficiency. Am J Respir Crit Care Med. JJ. High titer autoantibodies to GM-CSF 2009 2006;174(5):571 580 – in patients with AML, CML and MDS 32. Nogee LM, Dunbar AE III, Wert SE, 10. Kröner C, Wittmann T, Reu S, et al. Lung are associated with active disease. Askin F, Hamvas A, Whitsett JA. A disease caused by ABCA3 mutations. Leukemia. 2008;22(4):783–790 mutation in the surfactant protein Thorax. 2017;72(3):213–220 22. Nakano M, Omae K, Tanaka A, C gene associated with familial 11. Thouvenin G, Abou Taam R, Flamein et al. Causal relationship between interstitial lung disease. N Engl J Med. F, et al. Characteristics of disorders indium compound and 2001;344(8):573–579 associated with genetic mutations of effects on the lungs. J Occup Health. 33. Kleinlein B, Griese M, Liebisch G, et al. . Arch Dis Child. 2009;51(6):513–521 Fatal neonatal respiratory failure in an 2010;95(6):449–454 23. Noguchi S, Eitoku M, Kiyosawa H, infant with congenital hypothyroidism 12. Kröner C, Reu S, Teusch V, et al. Suganuma N. Fibrotic gene expression due to haploinsufficiency of Genotype alone does not predict the coexists with alveolar proteinosis the NKX2-1 gene: alteration of clinical course of SFTPC deficiency in early indium lung. Inhal Toxicol. pulmonary surfactant homeostasis. in paediatric patients. Eur Respir J. 2016;28(9):421–428 Arch Dis Child Fetal Neonatal Ed. 2011;96(6):F453 F456 2015;46(1):197–206 24. Omae K, Nakano M, Tanaka A, Hirata – 13. Hamvas A, Deterding RR, Wert SE, et al. M, Hamaguchi T, Chonan T. Indium 34. Wir tz HR, Dobbs LG. Calcium Heterogeneous pulmonary phenotypes lung–case reports and epidemiology. mobilization and associated with mutations in the Int Arch Occup Environ Health. after one mechanical stretch thyroid transcription factor gene NKX2- 2011;84(5):471–477 of lung epithelial cells. Science. 1990;250(4985):1266 1269 1. Chest. 2013;144(3):794–804 25. Nei T, Urano S, Itoh Y, et al. Light chain – 14. Hildebrandt J, Yalcin E, Bresser HG, (κ/λ) ratio of GM-CSF autoantibodies 35. W right JR, Dobbs LG. Regulation of et al. Characterization of CSF2RA is associated with disease severity pulmonary surfactant secretion mutation related juvenile pulmonary in autoimmune pulmonary and clearance. Annu Rev Physiol. alveolar proteinosis. Orphanet J Rare alveolar proteinosis. Clin Immunol. 1991;53:395–414 Dis. 2014;9(1):171 2013;149(3):357–364 36. Griese M, Gobran LI, Rooney SA. 15. Bonella F, Bauer PC, Griese M, 26. Arai T, Hamano E, Inoue Y, et al. Ontogeny of surfactant secretion Wessendorf TE, Guzman J, Costabel Serum neutralizing capacity of in type II pneumocytes from fetal, U. Wash-out kinetics and efficacy GM-CSF reflects disease severity in newborn, and adult rats. Am J Physiol. of a modified lavage technique for a patient with pulmonary alveolar 1992;262(3 pt 1):L337–L343 alveolar proteinosis. Eur Respir J. proteinosis successfully treated 37. Young LR, Deutsch GH, Bokulic RE, 2012;40(6):1468–1474 with inhaled GM-CSF. Respir Med. Brody AS, Nogee LM. A mutation in 16. Bonella F, Bauer PC, Griese M, Ohshimo 2004;98(12):1227–1230 TTF1/NKX2.1 is associated with familial S, Guzman J, Costabel U. Pulmonary 27. Griese M, Ripper J, Sibbersen A, et al. neuroendocrine cell hyperplasia of alveolar proteinosis: new insights from Long-term follow-up and treatment of infancy. Chest. 2013;144(4):1199–1206

Downloaded from www.aappublications.org/news by guest on October 1, 2021 PEDIATRICS Volume 140, number 2, August 2017 11 38. T redano M, Griese M, Brasch F, et al. alveolar proteinosis and its cytokine combination. Respir Med Case Rep. Mutation of SFTPC in infantile profile. BMC Pulm Med. 2017;17(1):40 2016;19:37–39 pulmonary alveolar proteinosis with 49. Griese M, Zarbock R, Costabel U, 60. Grunebaum E, Cutz E, Roifman CM. or without fibrosing lung disease. Am J et al. GATA2 deficiency in children Pulmonary alveolar proteinosis in Med Genet A. 2004;126A(1):18–26 and adults with severe pulmonary patients with adenosine deaminase 39. Rober tson B, Curstedt T, Herting E, alveolar proteinosis and hematologic deficiency.J Allergy Clin Immunol. Sun B, Akino T, Schäfer KP. Alveolar- disorders. BMC Pulm Med. 2015; 2012;129(6):1588–1593 to-vascular leakage of surfactant 15:87 61. Punatar AD, Kusne S, Blair JE, protein A in ventilated immature 50. Ishii H, Tazawa R, Kaneko C, et al. Seville MT, Vikram HR. Opportunistic newborn rabbits. Biol Neonate. Clinical features of secondary infections in patients with pulmonary 1995;68(3):185–190 pulmonary alveolar proteinosis: pre- alveolar proteinosis. J Infect. 40. Griese M, Beck J, Feuerhake mortem cases in Japan. Eur Respir 2012;65(2):173–179 F. Surfactant lipid uptake and J. 2011;37(2):465–468 62. Su KC, Lay SL, Perng RP, Chang SC, metabolism by neonatal and adult 51. Inaba H, Jenkins JJ, McCarville MB, Chen YM. Lung cancer may develop type II pneumocytes. Am J Physiol. et al. Pulmonary alveolar proteinosis subsequently or coincidently with 1999;277(5 pt 1):L901–L909 in pediatric leukemia. Pediatr Blood pulmonary alveolar proteinosis. Lung 41. Hadchouel A, Wieland T, Griese M, et al. Cancer. 2008;51(1):66–70 Cancer. 2007;58(1):144–148 Biallelic mutations of methionyl-tRNA 52. Ohnishi T, Yamada G, Shijubo N, 63. Campo I, Luisetti M, Griese M, et al; synthetase cause a specific type et al. Secondary pulmonary alveolar WLL International Study Group. A of pulmonary alveolar proteinosis proteinosis associated with global survey on whole lung lavage in prevalent on Réunion Island. Am J Hum myelodysplastic syndrome. Intern Med. pulmonary alveolar proteinosis. Chest. Genet. 2015;96(5):826–831 2003;42(2):187–190 2016;150(1):251–253 42. Carey B, Trapnell BC. The molecular 53. Spinner MA, Sanchez LA, Hsu AP, et al. 64. Campo I, Luisetti M, Griese M, et al; WLL basis of pulmonary alveolar GATA2 deficiency: a protean disorder International Study Group. Whole lung proteinosis. Clin Immunol. of hematopoiesis, lymphatics, and lavage therapy for pulmonary alveolar 2010;135(2):223–235 immunity. Blood. 2014;123(6):809–821 proteinosis: a global survey of current 43. Kitamura T, Tanaka N, Watanabe J, 54. Ogier de Baulny H, Schiff M, Dionisi-Vici practices and procedures. Orphanet et al. Idiopathic pulmonary alveolar C. Lysinuric protein intolerance J Rare Dis. 2016;11(1):115 proteinosis as an autoimmune (LPI): a multi organ disease by far 65. Wilson CA, Wilmshurst SL, Black AE. disease with neutralizing antibody more complex than a classic urea Anesthetic techniques to facilitate against granulocyte/macrophage cycle disorder. Mol Genet Metab. lung lavage for pulmonary alveolar colony-stimulating factor. J Exp Med. 2012;106(1):12–17 proteinosis in children-new airway 1999;190(6):875 880 – 55. Ceruti M, Rodi G, Stella GM, et al. techniques and a review of the 44. Costabel U, Guzman J. Pulmonary Successful whole lung lavage in literature. Paediatr Anaesth. alveolar proteinosis: a new pulmonary alveolar proteinosis 2015;25(6):546–553 autoimmune disease. Sarcoidosis secondary to lysinuric protein 66. Reiter K, Schoen C, Griese M, Nicolai Vasc Diffuse Lung Dis. 2005;22(suppl intolerance: a case report. Orphanet T. Whole-lung lavage in infants and 1):S67–S73 J Rare Dis. 2007;2:14 children with pulmonary alveolar 45. Mar tinez-Moczygemba M, Doan ML, 56. Valimahamed-Mitha S, Berteloot L, proteinosis. Paediatr Anaesth. Elidemir O, et al. Pulmonary alveolar Ducoin H, Ottolenghi C, de Lonlay P, de 2010;20(12):1118–1123 proteinosis caused by deletion of Blic J. Lung involvement in children 67. Parker LA, Novotny DB. Recurrent the GM-CSFRalpha gene in the X with lysinuric protein intolerance. alveolar proteinosis following chromosome pseudoautosomal J Inherit Metab Dis. 2015;38(2):257–263 double lung transplantation. Chest. region 1. J Exp Med. 57. Enaud L, Hadchouel A, Coulomb A, et 1997;111(5):1457–1458 2008;205(12):2711 2716 – al. Pulmonary alveolar proteinosis in 68. Takaki M, Tanaka T, Komohara Y, et al. 46. Notarangelo LD, Pessach I. Out of children on La Réunion Island: a new Recurrence of pulmonary alveolar breath: GM-CSFRalpha mutations inherited disorder? Orphanet J Rare proteinosis after bilateral lung disrupt surfactant homeostasis. J Exp Dis. 2014;9:85 transplantation in a patient with a Med. 2008;205(12):2693 2697 – 58. Griese M, Brasch F, Aldana VR, et al. nonsense mutation in CSF2RB. Respir 47. Tanaka T, Motoi N, Tsuchihashi Y, et al. in Niemann- Med Case Rep. 2016;19:89–93 Adult-onset hereditary pulmonary Pick type C2 is caused by pulmonary 69. Bonella F, Campo I. Pulmonary alveolar proteinosis caused by a alveolar proteinosis. Clin Genet. alveolar proteinosis. Pneumologia. single-base deletion in CSF2RB. J Med 2010;77(2):119–130 2014;63(3):144, 147–155 Genet. 2011;48(3):205–209 59. Sideris GA, Josephson M. Pulmonary 70. Campo I, Mariani F, Rodi G, et al. 48. Ito M, Nakagome K, Ohta H, et al. alveolar proteinosis and Niemann Assessment and management of Elderly-onset hereditary pulmonary Pick disease type B: an unexpected pulmonary alveolar proteinosis in a

Downloaded from www.aappublications.org/news by guest on October 1, 2021 12 Griese reference center. Orphanet J Rare Dis. in children and adults. Respir Res. 88. Ishii H, Seymour JF, Tazawa R, 2013;8:40 2005;6:138 et al. Secondary pulmonary 71. Papiris SA, Tsirigotis P, Kolilekas L, 80. Reuner t J, Lotz-Havla AS, Polo G, et al. alveolar proteinosis complicating et al. Long-term inhaled granulocyte Niemann-Pick type C-2 disease: myelodysplastic syndrome results macrophage-colony-stimulating factor identification by analysis of plasma in worsening of prognosis: a retrospective cohort study in Japan. in autoimmune pulmonary alveolar cholestane-3β,​5α,​6β-triol and further proteinosis: effectiveness, safety, insight into the clinical phenotype. BMC Pulm Med. 2014;14(1):37 and lowest effective dose. Clin Drug JIMD Rep. 2015;23:17–26 89. Barilli A, Rotoli BM, Visigalli R, et al. In Investig. 2014;34(8):553 564 Lysinuric Protein Intolerance system – 81. van Hoorn J, Brouwers A, Griese M, y+L activity is defective in monocytes 72. Tazawa R, Trapnell BC, Inoue Y, et al. Kramer B. Successful weaning from and in GM-CSF-differentiated Inhaled granulocyte/macrophage- in a patient macrophages. Orphanet J Rare Dis. colony stimulating factor as therapy with surfactant protein C deficiency 2010;5(1):32 for pulmonary alveolar proteinosis. presenting with severe neonatal Am J Respir Crit Care Med. respiratory distress. BMJ Case Rep. 90. Tanner LM, Kurko J, Tringham M, et al. 2010;181(12):1345–1354 2014;2014(1):bcr2013203053 Inhaled sargramostim induces resolution of pulmonary alveolar 73. Yamamoto H, Yamaguchi E, Agata H, 82. Tsai WC, Lewis D, Nasr SZ, Hirschl proteinosis in lysinuric protein et al. A combination therapy of RB. Liquid ventilation in an infant intolerance [published online ahead of whole lung lavage and GM-CSF with pulmonary alveolar proteinosis. print October 26, 2016]. JIMD Rep. doi:​ inhalation in pulmonary alveolar Pediatr Pulmonol. 1998;26(4): 10.​1007/​8904_​2016_​15 proteinosis. Pediatr Pulmonol. 283–286 2008;43(8):828–830 91. Gallagher J, Adams J, Hintermeyer 83. Cho K, Nakata K, Ariga T, et al. M, et al. X-linked hyper IgM syndrome 74. Kavuru MS, Bonfield TL, Thomassen MJ. Successful treatment of congenital presenting as pulmonary alveolar Plasmapheresis, GM-CSF, and alveolar pulmonary alveolar proteinosis proteinosis. J Clin Immunol. proteinosis. Am J Respir Crit Care with intravenous immunoglobulin 2016;36(6):564–570 Med. 2003;167(7):1036, author reply G administration. Respirology. 92. Patiroglu T, Akyildiz B, Patiroglu TE, 1036–1037 2006;11(suppl):S74 S77 – Gulmez IY. Recurrent pulmonary 75. Luisetti M, Rodi G, Perotti C, et al. 84. Wittmann T, Schindlbeck U, Höppner alveolar proteinosis secondary Plasmapheresis for treatment of S, et al. Tools to explore ABCA3 to agammaglobulinemia. Pediatr pulmonary alveolar proteinosis. Eur mutations causing interstitial Pulmonol. 2008;43(7):710–713 Respir J. 2009;33(5):1220–1222 lung disease. Pediatr Pulmonol. 93. Edwards C, Primhak R, Cohen MC. 76. Amital A, Dux S, Shitrit D, Shpilberg O, 2016;51(12):1284–1294 Pulmonary alveolar proteinosis Kramer MR. Therapeutic effectiveness 85. Cordonnier C, Fleury-Feith J, Escudier associated with Epstein-Barr of rituximab in a patient with E, Atassi K, Bernaudin JF. Secondary virus infection. Eur Respir J. unresponsive autoimmune pulmonary alveolar proteinosis is a reversible 2010;36(5):1214–1216 alveolar proteinosis. Thorax. cause of respiratory failure in 94. Narotzky S, Kennedy CC, Maldonado 2010;65(11):1025–1026 leukemic patients. Am J Respir Crit F. An unusual cause of respiratory 77. Malur A, Kavuru MS, Marshall I, et al. Care Med. 1994;149(3 pt 1):788–794 failure in a 25-year-old heart and Rituximab therapy in pulmonary 86. Tabata S, Shimoji S, Murase K, et al. lung transplant recipient. Chest. alveolar proteinosis improves alveolar Successful allogeneic bone marrow 2015;147(5):e185–e188 macrophage lipid homeostasis. Respir transplantation for myelodysplastic 95. Wardwell NR Jr, Miller R, Ware LB. Res. 2012;13:46 syndrome complicated by severe Pulmonary alveolar proteinosis 78. Nagasawa J, Kurasawa K, Hanaoka R. pulmonary alveolar proteinosis. Int associated with a disease-modifying Rituximab improved systemic lupus J Hematol. 2009;90(3):407–412 antirheumatoid arthritis drug. erythematosus-associated pulmonary 87. Xue Y, Han Y, Li T, et al. Pulmonary Respirology. 2006;11(5):663–665 alveolar proteinosis without alveolar proteinosis as a 96. Masuko H, Hizawa N, Chonan T, decreasing anti-GM-CSF antibody terminal complication in a case et al. Indium-tin oxide does not induce levels. Lupus. 2016;25(7):783 784 – of myelodysplastic syndrome GM-CSF autoantibodies. Am J Respir 79. Paschen C, Reiter K, Stanzel F, Teschler with idic(20q-). Acta Haematol. Crit Care Med. 2011;184(6):741, author H, Griese M. Therapeutic lung lavages 2010;123(1):55–58 reply 741–742

Downloaded from www.aappublications.org/news by guest on October 1, 2021 PEDIATRICS Volume 140, number 2, August 2017 13 Pulmonary Alveolar Proteinosis: A Comprehensive Clinical Perspective Matthias Griese Pediatrics 2017;140; DOI: 10.1542/peds.2017-0610 originally published online July 20, 2017;

Updated Information & including high resolution figures, can be found at: Services http://pediatrics.aappublications.org/content/140/2/e20170610 References This article cites 94 articles, 20 of which you can access for free at: http://pediatrics.aappublications.org/content/140/2/e20170610#BIBL Subspecialty Collections This article, along with others on similar topics, appears in the following collection(s): Pulmonology http://www.aappublications.org/cgi/collection/pulmonology_sub Respiratory Tract http://www.aappublications.org/cgi/collection/respiratory_tract_sub Permissions & Licensing Information about reproducing this article in parts (figures, tables) or in its entirety can be found online at: http://www.aappublications.org/site/misc/Permissions.xhtml Reprints Information about ordering reprints can be found online: http://www.aappublications.org/site/misc/reprints.xhtml

Downloaded from www.aappublications.org/news by guest on October 1, 2021 Pulmonary Alveolar Proteinosis: A Comprehensive Clinical Perspective Matthias Griese Pediatrics 2017;140; DOI: 10.1542/peds.2017-0610 originally published online July 20, 2017;

The online version of this article, along with updated information and services, is located on the World Wide Web at: http://pediatrics.aappublications.org/content/140/2/e20170610

Pediatrics is the official journal of the American Academy of Pediatrics. A monthly publication, it has been published continuously since 1948. Pediatrics is owned, published, and trademarked by the American Academy of Pediatrics, 345 Park Avenue, Itasca, Illinois, 60143. Copyright © 2017 by the American Academy of Pediatrics. All rights reserved. Print ISSN: 1073-0397.

Downloaded from www.aappublications.org/news by guest on October 1, 2021