1
|
Tito Rodriguez PR, Mehta D, Subhan M, Yadav RP, Yousofzai BS, Al-Najjar EH, Bibi R, Idries M, Singh A, Adnan M. Evolving Horizons in Pediatric Leukemia: Novel Insights, Challenges, and the Journey Ahead. Cureus 2024; 16:e67480. [PMID: 39310608 PMCID: PMC11415937 DOI: 10.7759/cureus.67480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/22/2024] [Indexed: 09/25/2024] Open
Abstract
Pediatric leukemia, encompassing acute lymphoblastic leukemia (ALL) and acute myeloid leukemia, remains a formidable challenge despite significant treatment advancements. This review examines recent developments in immunotherapy, chemotherapy, and bone marrow transplantation for pediatric leukemia through a comprehensive analysis of recent literature, focusing on critical studies and clinical trials. Immunotherapy, including monoclonal antibodies, such as blinatumomab and inotuzumab ozogamicin, and chimeric antigen receptor T-cell therapies, such as tisagenlecleucel and brexucabtagene autoleucel, have demonstrated promising results in relapsed or refractory B-cell ALL (B-ALL), achieving notable remission rates with manageable side effects. Chemotherapy continues to be the primary treatment, utilizing multiphase regimens tailored to individual risk profiles. Bone marrow transplantation, especially allogeneic stem cell transplantation, offers potential cures for high-risk or relapsed cases, though it poses risks including graft-versus-host disease and infections. Despite these advancements, treatment resistance, toxicity, and accessibility persist. This review also discusses the long-term outcomes among pediatric leukemia survivors, focusing on late-onset side effects associated with treatments such as chemotherapy and bone marrow transplantation, encompassing secondary malignancies, organ dysfunction, and neurocognitive impacts. Ongoing research and clinical trials are crucial to refine these therapies, enhance their efficacy, and reduce adverse effects, ultimately improving young patients' survival and quality of life.
Collapse
Affiliation(s)
| | - Deepalee Mehta
- Internal Medicine, Bharati Vidyapeeth Medical College, Sangli, Sangli, IND
| | - Muhammad Subhan
- Medicine, Allama Iqbal Medical College, Jinnah Hospital, Lahore, PAK
| | | | | | | | - Ruqiya Bibi
- Medicine, Allama Iqbal Medical College, Jinnah Hospital, Lahore, PAK
| | - Mohamed Idries
- Biochemistry, St. George's University School of Medicine, St. George's, GRD
| | - Atinder Singh
- Medicine, World College of Medical Sciences and Research Hospital, Gurugram, IND
| | - Muhammad Adnan
- Pediatrics, Lady Reading Hospital, Peshawar, PAK
- Pediatrics, Khyber Medical College, Peshawar, PAK
| |
Collapse
|
2
|
Li Q, Ren X, Wang Y, Xin X. CircRNA: a rising star in leukemia. PeerJ 2023; 11:e15577. [PMID: 37431465 PMCID: PMC10329819 DOI: 10.7717/peerj.15577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 05/25/2023] [Indexed: 07/12/2023] Open
Abstract
Non-coding RNA are a class of RNA that lack the potential to encode proteins. CircRNAs, generated by a post-splicing mechanism, are a newly discovered type of non-coding RNA with multi-functional covalent loop structures. CircRNAs may play an important role in the occurrence and progression of tumors. Research has shown that circRNAs are aberrantly expressed in various types of human cancers, including leukemia. In this review, we summarize the expression and function of circRNAs and their impact on different types of leukemia. We also illustrate the function of circRNAs on immune modulation and chemoresistance in leukemia and their impact on its diagnosis and prognosis. Herein, we provide an understanding of recent advances in research that highlight the importance of circRNAs in proliferation, apoptosis, migration, and autophagy in different types of leukemia. Furthermore, circRNAs make an indispensable difference in the modulation of the immunity and chemoresistance of leukemia. Increasing evidence suggests that circRNAs may play a vital role in the diagnostic and prognostic markers of leukemia because of their prominent properties. More detailed preclinical studies on circRNAs are needed to explore effective ways in which they can serve as biomarkers for the diagnosis and prognosis of leukemia in vivo.
Collapse
Affiliation(s)
- Qianan Li
- College of Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Xinxin Ren
- College of Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Ying Wang
- College of Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Xiaoru Xin
- College of Life Sciences, Zhejiang Normal University, Jinhua, China
| |
Collapse
|
3
|
Domingo-Reinés J, Martínez-Navajas G, Montes R, Lamolda M, Simón I, Castaño J, Ríos-Pelegrina R, Lopez-Hidalgo JL, García del Moral R, Marchal JA, Real PJ, Ramos-Mejía V. Generation of a H9 Clonal Cell Line With Inducible Expression of NUP98-KDM5A Fusion Gene in the AAVS1 Safe Harbor Locus. Front Cell Dev Biol 2022; 10:846092. [PMID: 35721502 PMCID: PMC9200071 DOI: 10.3389/fcell.2022.846092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 05/12/2022] [Indexed: 11/25/2022] Open
Abstract
Pediatric acute myeloid leukemia (AML) is a rare and heterogeneous disease that remains the major cause of mortality in children with leukemia. To improve the outcome of pediatric AML we need to gain knowledge on the biological bases of this disease. NUP98-KDM5A (NK5A) fusion protein is present in a particular subgroup of young pediatric patients with poor outcome. We report the generation and characterization of human Embryonic Stem Cell (hESC) clonal lines with inducible expression of NK5A. Temporal control of NK5A expression during hematopoietic differentiation from hESC will be critical for elucidating its participation during the leukemogenic process.
Collapse
Affiliation(s)
- Joan Domingo-Reinés
- GENYO, Centre for Genomics and Oncological Research Pfizer-University of Granada-Andalusian Regional Government, Granada, Spain
| | - Gonzalo Martínez-Navajas
- GENYO, Centre for Genomics and Oncological Research Pfizer-University of Granada-Andalusian Regional Government, Granada, Spain
- Faculty of Science, Department of Biochemistry and Molecular Biology I, University of Granada, Granada, Spain
| | - Rosa Montes
- GENYO, Centre for Genomics and Oncological Research Pfizer-University of Granada-Andalusian Regional Government, Granada, Spain
- Faculty of Sciences, Department of Cell Biology, University of Granada, Granada, Spain
| | - Mar Lamolda
- GENYO, Centre for Genomics and Oncological Research Pfizer-University of Granada-Andalusian Regional Government, Granada, Spain
| | - Iris Simón
- GENYO, Centre for Genomics and Oncological Research Pfizer-University of Granada-Andalusian Regional Government, Granada, Spain
- Faculty of Science, Department of Biochemistry and Molecular Biology I, University of Granada, Granada, Spain
| | - Julio Castaño
- Advanced Cell Therapy Service, Banc de Sang I Teixits, Edifici Dr. Frederic Duran I Jordà, Barcelona, Spain
| | - Rosa Ríos-Pelegrina
- Unidad de Anatomía Patológica, Hospital Universitario Clínico San Cecilio, Granada, Spain
- Instituto de Investigación Biosanitaria Ibs.GRANADA, Granada, Spain
| | - Javier Luis Lopez-Hidalgo
- Unidad de Anatomía Patológica, Hospital Universitario Clínico San Cecilio, Granada, Spain
- Instituto de Investigación Biosanitaria Ibs.GRANADA, Granada, Spain
| | - Raimundo García del Moral
- Unidad de Anatomía Patológica, Hospital Universitario Clínico San Cecilio, Granada, Spain
- Instituto de Investigación Biosanitaria Ibs.GRANADA, Granada, Spain
| | - Juan A. Marchal
- Instituto de Investigación Biosanitaria Ibs.GRANADA, Granada, Spain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, Granada, Spain
- Faculty of Medicine, Department of Human Anatomy and Embryology, University of Granada, Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, Granada, Spain
| | - Pedro J. Real
- GENYO, Centre for Genomics and Oncological Research Pfizer-University of Granada-Andalusian Regional Government, Granada, Spain
- Faculty of Science, Department of Biochemistry and Molecular Biology I, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria Ibs.GRANADA, Granada, Spain
| | - Verónica Ramos-Mejía
- GENYO, Centre for Genomics and Oncological Research Pfizer-University of Granada-Andalusian Regional Government, Granada, Spain
- *Correspondence: Verónica Ramos-Mejía,
| |
Collapse
|
4
|
The Molecular Subtype of Adult Acute Lymphoblastic Leukemia Samples Determines the Engraftment Site and Proliferation Kinetics in Patient-Derived Xenograft Models. Cells 2022; 11:cells11010150. [PMID: 35011712 PMCID: PMC8750004 DOI: 10.3390/cells11010150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/30/2021] [Accepted: 12/31/2021] [Indexed: 12/28/2022] Open
Abstract
In acute lymphoblastic leukemia (ALL), conventional cell lines do not recapitulate the clonal diversity and microenvironment. Orthotopic patient-derived xenograft models (PDX) overcome these limitations and mimic the clinical situation, but molecular stability and engraftment patterns have not yet been thoroughly assessed. We herein describe and characterize the PDX generation in NSG mice. In vivo tumor cell proliferation, engraftment and location were monitored by flow cytometry and bioluminescence imaging. Leukemic cells were retransplanted for up to four passages, and comparative analyses of engraftment pattern, cellular morphology and genomic hotspot mutations were conducted. Ninety-four percent of all samples were successfully engrafted, and the xenograft velocity was dependent on the molecular subtype, outcome of the patient and transplantation passage. While BCR::ABL1 blasts were located in the spleen, KMT2A-positive cases had higher frequencies in the bone marrow. Molecular changes appeared in most model systems, with low allele frequency variants lost during primary engraftment. After the initial xenografting, however, the PDX models demonstrated high molecular stability. This protocol for reliable ALL engraftment demonstrates variability in the location and molecular signatures during serial transplantation. Thorough characterization of experimentally used PDX systems is indispensable for the correct analysis and valid data interpretation of preclinical PDX studies.
Collapse
|
5
|
Cytogenetic risk groups for childhood acute myeloid leukemia based on survival analysis in a cancer referral hospital from Perú. ACTA ACUST UNITED AC 2021; 41:302-313. [PMID: 34214271 PMCID: PMC8382291 DOI: 10.7705/biomedica.5747] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Indexed: 12/27/2022]
Abstract
Introduction: Acute myeloid leukemia is a heterogeneous disorder characterized by immature myeloid cell proliferation. Cytogenetic analysis has revealed the presence of chromosomal aberrations important to patient prognosis.
Objective: To determine cytogenetic risk groups of pediatric patients with acute myeloid leukemia according to overall survival.
Materials and methods: In this cross-sectional observational study, the clinical records of pediatric patients diagnosed with de novo acute myeloid leukemia admitted to the Instituto Nacional de Enfermedades Neoplásicas between 2001 and 2011 with cytogenetic analysis of bone marrow were included. Cytogenetic risk groups were established according to the criteria of the Medical Research Council. Overall survival curves were generated with the Kaplan-Meier method and compared using the Mantel-Cox test and Cox regression with the software R, version 3.3.2.
Results: A total of 130 patients were included, 68 males (52.3%) and 62 females (47.7%), most of them with subtype M2 (33%). The average age was 7.7 years (range: 0-15 years). Chromosomal aberrations were observed in 60.8% of the patients, the most frequent of which was the translocation t(8;21). According to the overall survival analysis, two cytogenetic risk groups were established: favorable and unfavorable.
Conclusion: Two groups of cytogenetic risk were determined: high (or unfavorable) and standard (favorable).
Collapse
|
6
|
A reporter system for enriching CRISPR/Cas9 knockout cells in technically challenging settings like patient models. Sci Rep 2021; 11:12649. [PMID: 34135367 PMCID: PMC8209181 DOI: 10.1038/s41598-021-91760-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 05/21/2021] [Indexed: 11/13/2022] Open
Abstract
CRISPR/Cas9 represents a valuable tool to determine protein function, but technical hurdles limit its use in challenging settings such as cells unable to grow in vitro like primary leukemia cells and xenografts derived thereof (PDX). To enrich CRISPR/Cas9-edited cells, we improved a dual-reporter system and cloned the genomic target sequences of the gene of interest (GOI) upstream of an out-of-frame fluorochrome which was expressed only upon successful gene editing. To reduce rounds of in vivo passaging required for PDX leukemia growth, targets of 17 GOI were cloned in a row, flanked by an improved linker, and PDX cells were lentivirally transduced for stable expression. The reporter enriched scarce, successfully gene-edited PDX cells as high as 80%. Using the reporter, we show that KO of the SRC-family kinase LYN increased the response of PDX cells of B precursor cell ALL towards Vincristine, even upon heterozygous KO, indicating haploinsufficiency. In summary, our reporter system enables enriching KO cells in technically challenging settings and extends the use of gene editing to highly patient-related model systems.
Collapse
|
7
|
Keinan N, Scharff Y, Goldstein O, Chamo M, Ilic S, Gazit R. Syngeneic leukemia models using lentiviral transgenics. Cell Death Dis 2021; 12:193. [PMID: 33602907 PMCID: PMC7893004 DOI: 10.1038/s41419-021-03477-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 01/31/2023]
Abstract
Animal models are necessary to study cancer and develop treatments. After decades of intensive research, effective treatments are available for only a few types of leukemia, while others are currently incurable. Our goal was to generate novel leukemia models in immunocompetent mice. We had achieved abilities for overexpression of multiple driving oncogenes simultaneously in normal primary cells, which can be transplanted and followed in vivo. Our experiments demonstrated the induction of primary malignant growth. Leukemia lines that model various types of leukemia, such as acute myeloid leukemia (AML) or chronic lymphocytic leukemia (CLL), were passaged robustly in congenic wild-type immunocompetent mice. These novel leukemia lines, which may complement previous models, offer the flexibility to generate tailored models of defined oncogenes of interest. The characterization of our leukemia models in immunocompetent animals can uncover the mechanisms of malignancy progression and offer a unique opportunity to stringently test anti-cancer chemotherapies.
Collapse
MESH Headings
- Animals
- Antimetabolites, Antineoplastic/pharmacology
- Cell Line, Tumor
- Cell Proliferation
- Cell Transformation, Viral
- Gene Expression Regulation, Leukemic
- Hematopoietic Stem Cells/immunology
- Hematopoietic Stem Cells/pathology
- Hematopoietic Stem Cells/virology
- Immunocompetence
- Lentivirus/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/virology
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/virology
- Mice, Inbred C57BL
- Mice, Transgenic
- Neoplasm Transplantation
- Oncogenes
- Transplantation, Isogeneic
- Vidarabine/analogs & derivatives
- Vidarabine/pharmacology
- Mice
Collapse
Affiliation(s)
- Nurit Keinan
- The Shraga Segal Department for Microbiology, Immunology, and Genetics, Faculty of Health Sciences; National Institute for Biotechnology in the Negev, the Ben-Gurion University of the Negev, Beer-Sheva, POB 84105, Israel
| | - Ye'ela Scharff
- The Shraga Segal Department for Microbiology, Immunology, and Genetics, Faculty of Health Sciences; National Institute for Biotechnology in the Negev, the Ben-Gurion University of the Negev, Beer-Sheva, POB 84105, Israel
| | - Oron Goldstein
- The Shraga Segal Department for Microbiology, Immunology, and Genetics, Faculty of Health Sciences; National Institute for Biotechnology in the Negev, the Ben-Gurion University of the Negev, Beer-Sheva, POB 84105, Israel
| | - Michael Chamo
- The Shraga Segal Department for Microbiology, Immunology, and Genetics, Faculty of Health Sciences; National Institute for Biotechnology in the Negev, the Ben-Gurion University of the Negev, Beer-Sheva, POB 84105, Israel
| | - Stefan Ilic
- The Shraga Segal Department for Microbiology, Immunology, and Genetics, Faculty of Health Sciences; National Institute for Biotechnology in the Negev, the Ben-Gurion University of the Negev, Beer-Sheva, POB 84105, Israel
| | - Roi Gazit
- The Shraga Segal Department for Microbiology, Immunology, and Genetics, Faculty of Health Sciences; National Institute for Biotechnology in the Negev, the Ben-Gurion University of the Negev, Beer-Sheva, POB 84105, Israel.
| |
Collapse
|
8
|
Sommerkamp P, Mercier FE, Wilkinson AC, Bonnet D, Bourgine PE. Engineering human hematopoietic environments through ossicle and bioreactor technologies exploitation. Exp Hematol 2021; 94:20-25. [PMID: 33278488 PMCID: PMC7879567 DOI: 10.1016/j.exphem.2020.11.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/23/2020] [Accepted: 11/29/2020] [Indexed: 01/16/2023]
Abstract
The bone marrow microenvironment contains cellular niches that maintain the pool of hematopoietic stem and progenitor cells and support hematopoietic maturation. Malignant hematopoietic cells also co-opt normal cellular interactions to promote their own growth and evade therapy. In vivo systems used to study human hematopoiesis have been developed through transplantation into immunodeficient mouse models. However, incomplete cross-compatibility between the murine stroma and transplanted human hematopoietic cells limits the rate of engraftment and the study of relevant interactions. To supplement in vivo xenotransplantation models, complementary strategies have recently been developed, including the use of three-dimensional human bone marrow organoids in vivo, generated from bone marrow stromal cells seeded onto osteo-inductive scaffolds, as well as the use of ex vivo bioreactor models. These topics were the focus of the Spring 2020 International Society for Experimental Hematology New Investigator webinar. We review here the latest advances in generating humanized hematopoietic organoids and how they allow for the study of novel microenvironmental interactions.
Collapse
Affiliation(s)
- Pia Sommerkamp
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
| | - François E Mercier
- Lady Davis Institute for Medical Research, Department of Medicine, McGill University, Montreal, Quebec, Canada.
| | - Adam C Wilkinson
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA
| | - Dominique Bonnet
- The Francis Crick Institute, Haematopoietic Stem Cell Laboratory, London, UK
| | - Paul E Bourgine
- Laboratory for Cell, Tissue, and Organ Engineering, Department of Clinical Sciences, Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Stem Cell Center, Lund University, Lund, Sweden
| |
Collapse
|
9
|
Liu WH, Mrozek-Gorska P, Wirth AK, Herold T, Schwarzkopf L, Pich D, Völse K, Melo-Narváez MC, Carlet M, Hammerschmidt W, Jeremias I. Inducible transgene expression in PDX models in vivo identifies KLF4 as a therapeutic target for B-ALL. Biomark Res 2020; 8:46. [PMID: 32944247 PMCID: PMC7493381 DOI: 10.1186/s40364-020-00226-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 09/07/2020] [Indexed: 02/08/2023] Open
Abstract
Background Clinically relevant methods are not available that prioritize and validate potential therapeutic targets for individual tumors, from the vast amount of tumor descriptive expression data. Methods We established inducible transgene expression in clinically relevant patient-derived xenograft (PDX) models in vivo to fill this gap. Results With this technique at hand, we analyzed the role of the transcription factor Krüppel-like factor 4 (KLF4) in B-cell acute lymphoblastic leukemia (B-ALL) PDX models at different disease stages. In competitive preclinical in vivo trials, we found that re-expression of wild type KLF4 reduced the leukemia load in PDX models of B-ALL, with the strongest effects being observed after conventional chemotherapy in minimal residual disease (MRD). A nonfunctional KLF4 mutant had no effect on this model. The re-expression of KLF4 sensitized tumor cells in the PDX model towards systemic chemotherapy in vivo. It is of major translational relevance that azacitidine upregulated KLF4 levels in the PDX model and a KLF4 knockout reduced azacitidine-induced cell death, suggesting that azacitidine can regulate KLF4 re-expression. These results support the application of azacitidine in patients with B-ALL as a therapeutic option to regulate KLF4. Conclusion Genetic engineering of PDX models allows the examination of the function of dysregulated genes like KLF4 in a highly clinically relevant translational context, and it also enables the selection of therapeutic targets in individual tumors and links their functions to clinically available drugs, which will facilitate personalized treatment in the future.
Collapse
Affiliation(s)
- Wen-Hsin Liu
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Marchioninistraße 25, 81377 Munich, Germany
| | - Paulina Mrozek-Gorska
- Research Unit Gene Vectors, Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Munich, Germany
| | - Anna-Katharina Wirth
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Marchioninistraße 25, 81377 Munich, Germany
| | - Tobias Herold
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Marchioninistraße 25, 81377 Munich, Germany.,Laboratory for Leukemia Diagnostics, Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Larissa Schwarzkopf
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Marchioninistraße 25, 81377 Munich, Germany
| | - Dagmar Pich
- Research Unit Gene Vectors, Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Munich, Germany
| | - Kerstin Völse
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Marchioninistraße 25, 81377 Munich, Germany
| | - M Camila Melo-Narváez
- Research Unit Gene Vectors, Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Munich, Germany
| | - Michela Carlet
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Marchioninistraße 25, 81377 Munich, Germany
| | - Wolfgang Hammerschmidt
- Research Unit Gene Vectors, Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Irmela Jeremias
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Marchioninistraße 25, 81377 Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.,Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig Maximilian University, Munich, Germany
| |
Collapse
|