Detailseite
Projekt Druckansicht

Genetisch korrigierte induziert pluripotente Stammzellen abgeleitete Gewebs-Makrophagen als innovative Therapie für Pulmonale Alveolar Proteinose (PAP)

Fachliche Zuordnung Hämatologie, Onkologie
Pneumologie,Thoraxchirurgie
Förderung Förderung von 2015 bis 2019
Projektkennung Deutsche Forschungsgemeinschaft (DFG) - Projektnummer 275410142
 
Erstellungsjahr 2019

Zusammenfassung der Projektergebnisse

The main aim of this proposal was to establish a disease modeling platform and a new therapeutic option for the rare congenital lung disease “Pulmonary Alveolar Proteinosis (PAP)”. As no long-term treatment for this disease is available, new and innovative strategies are highly desirable. Because alveolar macrophages (immune cell in the bronchioalveolar space) are the main effector cells of PAP, we established a macrophage production pipeline, which is based on induced pluripotent stem cell technology. This newly generated pipeline allows for the continuous generation of macrophages (and other blood cells) over a time period of several months. Using this technology, we were able gain new insights into PAP and to establish a new gene therapy approach for this disease. As a next step, we evaluated the therapeutic efficacy of intra-pulmonary macrophage transplantation. Here, we used two different mouse models for PAP and have investigated the pre-clinical outcome of murine and human iPSC-derived macrophages two and six months post intra-pulmonary transplantation. Using the afore mentioned technology, we transplanted 2-4x106 macrophages into PAP mice and could observe improved disease parameters in transplanted mice compared to untreated contols. Notably, macrophages transplanted directly into the lungs of PAP mice showed local engraftment and could not be detected in other organs e.g. liver, brain, spleen or bone marrow. Moreover, transplanted macrophages could even be detected up to six-months post transplantation, indicating successful engraftment and long-term persistence of transplanted cells. Indeed, using transcriptome analysis, we observed an in vivo adaption potential of iPSC-derived macrophages within two-months post transplantation. Here, human iPSC-derived macrophages showed a similar transcriptome profile to primary human alveolar macrophages, which was very much distinct from iPSC-macrophages before transplantation, indicating in in vivo adaption of transplanted cells. Given the high medical implication of iPSC technology, we have been the first who have used mature iPSC-derived blood cells to improve the disease symptoms in vivo. Along this line, we also describe a profound stem cell-like phenotype of mature iPSC-derived macrophages and highlight the ability of iPSC-derived macrophages to self-maintain and adapt towards an alveolar-macrophage like phenotype within in the lung microenvironment. These newly introduced concepts were also highlighted by different reports for the public (“Murine iPSC-macrophage transplantation” and “Heilung mit eigenen Zellen in Aussicht”). Of note, the newly introduced macrophage transplantation concept has also broad application for other disease entities. Macrophages can be found in various tissues and malfunction of macrophages has been associated with different disease entities. Thus, the transplantation of macrophages, local engraftment and adaptation potential may also be used for other disorders affecting the lung, brain, liver or gut. Current efforts in the lab are investigating macrophage transplantation for Cystic Fibrosis (lung) or Inflammatory Bowel Disease (gut). To achieve this aim and to drive macrophage transplantation into clinical translation, the afore mentioned macrophage production platform has been further advanced to be compatible with industry compatible stirred tank bioreactors. Using this technology, we are now able to generate human iPSC-derived macrophages in scalable quantities, allowing us to investigate completely new forms of immunotherapies. Similarly, this technology has been also highlighted by different reports for the public (“Biotechnologie: Immunzellen aus dem Bioreaktor” and “Makrophagen hergestellt: Blut aus dem Bioreaktor gegen Infektionen”). In summary, we here introduce human iPSC-derived macrophages for PAP and lay the foundation for other macrophage-related disease entities and beyond.

Projektbezogene Publikationen (Auswahl)

  • „Murine iPSC-derived macrophages as a tool for disease modeling of hereditary Pulmonary Alveolar Proteinosis due to Csf2rb deficiency“. Stem Cell Reports. 2016 Aug 9;7(2):292-305
    Mucci A, Kunkiel J, Suzuki T, Brennig S, Glage S, Kühnel MP, Ackermann M, Happle C, Kuhn A, Schambach A, Trapnell BC, Hansen G, Moritz T, Lachmann N
    (Siehe online unter https://doi.org/10.1016/j.stemcr.2016.06.011)
  • “Ex vivo Generation of Genetically Modified Macrophages from Human Induced Pluripotent Stem Cells”. Transfus Med Hemother 2017 Jun;44(3):135-142
    Ackermann M, Kuhn A, Kunkiel J, Merkert S, Martin U, Moritz T, Lachmann N
    (Siehe online unter https://doi.org/10.1159/000477129)
  • “Function and Safety of Lentivirus-Mediated Gene Transfer for CSF2RA-Deficiency”. Hum Gene Ther Methods. 2017 Dec;28(6):318-329
    Hetzel M, Suzuki T, Hashtchin AR, Arumugam P, Carey B, Schwabbauer M, Kuhn A, Meyer J, Schambach A, Van Der Loo J, Moritz T, Trapnell BC, Lachmann N
    (Siehe online unter https://doi.org/10.1089/hgtb.2017.092)
  • “TALEN-mediated functional correction of human iPSC-derived macrophages in context of hereditary pulmonary alveolar proteinosis.” Sci Rep. 2017 Nov 9;7(1):15195
    Kuhn A, Ackermann M, Mussolino C, Cathomen T, Lachmann N, Moritz T
    (Siehe online unter https://doi.org/10.1038/s41598-017-14566-8)
  • “Pulmonary transplantation of human iPSC-derived macrophages ameliorates hereditary lung disease”. Am J Respir Crit Care Med. 2018 Apr 13
    Happle C, Lachmann N, Ackermann M, Wetzke M, Mirenska A, Göhring G, Thomay K, Mucci A, Glomb T, Suzuki T, Glage S, Dittrich-Breiholz O, Trapnell B, Moritz T, Hansen G
    (Siehe online unter https://doi.org/10.1164/rccm.201708-1562OC)
  • „Bioreactor-based mass production of human iPSC-derived macrophages enables immunotherapies against bacterial airway infections.“ Nature Communications. 2018 Nov 30;9(1):5088
    Ackermann M, Kempf H, Hetzel M, Hesse C, Hashtchin AR, Brinkert K, Schott JW, Haake K, Kühnel MP, Glage S, Figueiredo C, Jonigk D, Sewald K, Schambach A, Wronski S, Moritz T, Martin U, Zweigerdt R, Munder A, Lachmann N
    (Siehe online unter https://doi.org/10.1038/s41467-018-07570-7)
  • „iPSC-Derived Macrophages Effectively Treat Pulmonary Alveolar Proteinosis in Csf2rb-Deficient Mice.“ Stem Cell Reports. 2018 Sep 11;11(3):696-710
    Mucci A, Lopez-Rodriguez E, Hetzel M, Liu S, Suzuki T, Happle C, Ackermann M, Kempf H, Hillje R, Kunkiel J, Janosz E, Brennig S, Glage S, Bankstahl JP, Dettmer S, Rodt T, Gohring G, Trapnell B, Hansen G, Trapnell C, Knudsen L, Lachmann N, Moritz T
    (Siehe online unter https://doi.org/10.1016/j.stemcr.2018.07.006)
  • „Towards the Clinical Translation of Induced Pluripotent Stem Cell-Derived Blood Cells-Ready for Take-Off“. Stem Cells Transl Med. 2019 Apr;8(4):332-339
    Haake K, Ackermann M, Lachmann N
    (Siehe online unter https://doi.org/10.1002/sctm.18-0134)
 
 

Zusatzinformationen

Textvergrößerung und Kontrastanpassung