1
|
Kowarsch F, Maurer-Granofszky M, Weijler L, Wödlinger M, Reiter M, Schumich A, Feuerstein T, Sala S, Nováková M, Faggin G, Gaipa G, Hrusak O, Buldini B, Dworzak MN. FCM marker importance for MRD assessment in T-cell acute lymphoblastic leukemia: An AIEOP-BFM-ALL-FLOW study group report. Cytometry A 2024; 105:24-35. [PMID: 37776305 DOI: 10.1002/cyto.a.24805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 08/07/2023] [Accepted: 09/18/2023] [Indexed: 10/02/2023]
Abstract
T-lineage acute lymphoblastic leukemia (T-ALL) accounts for about 15% of pediatric and about 25% of adult ALL cases. Minimal/measurable residual disease (MRD) assessed by flow cytometry (FCM) is an important prognostic indicator for risk stratification. In order to assess the MRD a limited number of antibodies directed against the most discriminative antigens must be selected. We propose a pipeline for evaluating the influence of different markers for cell population classification in FCM data. We use linear support vector machine, fitted to each sample individually to avoid issues with patient and laboratory variations. The best separating hyperplane direction as well as the influence of omitting specific markers is considered. Ninety-one bone marrow samples of 43 pediatric T-ALL patients from five reference laboratories were analyzed by FCM regarding marker importance for blast cell identification using combinations of eight different markers. For all laboratories, CD48 and CD99 were among the top three markers with strongest contribution to the optimal hyperplane, measured by median separating hyperplane coefficient size for all samples per center and time point (diagnosis, Day 15, Day 33). Based on the available limited set tested (CD3, CD4, CD5, CD7, CD8, CD45, CD48, CD99), our findings prove that CD48 and CD99 are useful markers for MRD monitoring in T-ALL. The proposed pipeline can be applied for evaluation of other marker combinations in the future.
Collapse
Affiliation(s)
- Florian Kowarsch
- Computer Vision Lab, Faculty of Informatics, Technical University of Vienna, Vienna, Austria
| | - Margarita Maurer-Granofszky
- Immunological Diagnostics, St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
- Labdia Labordiagnostik GmbH, Vienna, Austria
| | - Lisa Weijler
- Computer Vision Lab, Faculty of Informatics, Technical University of Vienna, Vienna, Austria
| | - Matthias Wödlinger
- Computer Vision Lab, Faculty of Informatics, Technical University of Vienna, Vienna, Austria
- Immunological Diagnostics, St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Michael Reiter
- Computer Vision Lab, Faculty of Informatics, Technical University of Vienna, Vienna, Austria
- Immunological Diagnostics, St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Angela Schumich
- Immunological Diagnostics, St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Tamar Feuerstein
- The Rina Zaizov Division of Pediatric Hematology-Oncology, Schneider's Children's Medical Center, Petah Tikva, Israel
| | - Simona Sala
- M. Tettamanti Foundation Research Center, Department of Pediatrics, University of Milano-Bicocca, Monza, Italy
| | - Michaela Nováková
- Department of Pediatric Haematology and Oncology, University Hospital Motol, Prague, Czech Republic
| | - Giovanni Faggin
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Maternal and Child Health Department, University of Padova, Padova, Italy
| | - Giuseppe Gaipa
- M. Tettamanti Foundation Research Center, Department of Pediatrics, University of Milano-Bicocca, Monza, Italy
| | - Ondrej Hrusak
- Department of Pediatric Haematology and Oncology, University Hospital Motol, Prague, Czech Republic
| | - Barbara Buldini
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Maternal and Child Health Department, University of Padova, Padova, Italy
- Advanced Diagnostics and Target Discovery in ALL, Fondazione istituto di Ricerca pediatrica Città della Speranza, Padova, Italy
| | - Michael N Dworzak
- Immunological Diagnostics, St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
- Labdia Labordiagnostik GmbH, Vienna, Austria
| |
Collapse
|
2
|
Tislevoll BS, Hellesøy M, Fagerholt OHE, Gullaksen SE, Srivastava A, Birkeland E, Kleftogiannis D, Ayuda-Durán P, Piechaczyk L, Tadele DS, Skavland J, Panagiotis B, Hovland R, Andresen V, Seternes OM, Tvedt THA, Aghaeepour N, Gavasso S, Porkka K, Jonassen I, Fløisand Y, Enserink J, Blaser N, Gjertsen BT. Early response evaluation by single cell signaling profiling in acute myeloid leukemia. Nat Commun 2023; 14:115. [PMID: 36611026 PMCID: PMC9825407 DOI: 10.1038/s41467-022-35624-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 12/13/2022] [Indexed: 01/09/2023] Open
Abstract
Aberrant pro-survival signaling is a hallmark of cancer cells, but the response to chemotherapy is poorly understood. In this study, we investigate the initial signaling response to standard induction chemotherapy in a cohort of 32 acute myeloid leukemia (AML) patients, using 36-dimensional mass cytometry. Through supervised and unsupervised machine learning approaches, we find that reduction of extracellular-signal-regulated kinase (ERK) 1/2 and p38 mitogen-activated protein kinase (MAPK) phosphorylation in the myeloid cell compartment 24 h post-chemotherapy is a significant predictor of patient 5-year overall survival in this cohort. Validation by RNA sequencing shows induction of MAPK target gene expression in patients with high phospho-ERK1/2 24 h post-chemotherapy, while proteomics confirm an increase of the p38 prime target MAPK activated protein kinase 2 (MAPKAPK2). In this study, we demonstrate that mass cytometry can be a valuable tool for early response evaluation in AML and elucidate the potential of functional signaling analyses in precision oncology diagnostics.
Collapse
Affiliation(s)
- Benedicte Sjo Tislevoll
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Monica Hellesøy
- Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Helse Bergen HF, Bergen, Norway
| | - Oda Helen Eck Fagerholt
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Stein-Erik Gullaksen
- Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Helse Bergen HF, Bergen, Norway
| | - Aashish Srivastava
- Genome Core Facility, Clinical Laboratory, K2 Haukeland University Hospital, Bergen, Norway
| | - Even Birkeland
- The Proteomics Facility of the University of Bergen (PROBE), University of Bergen, Bergen, Norway
| | - Dimitrios Kleftogiannis
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Science, University of Bergen, Bergen, Norway.,Centre for Cancer Biomarkers and Computational Biology Unit, Department of Informatics, University of Bergen, Bergen, Norway
| | - Pilar Ayuda-Durán
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Montebello, 0379, Oslo, Norway.,Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0318, Oslo, Norway
| | - Laure Piechaczyk
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Montebello, 0379, Oslo, Norway.,Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0318, Oslo, Norway.,Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Dagim Shiferaw Tadele
- Department of Molecular Genetics, Division of Laboratory Medicine, Oslo University Hospital, Oslo, Norway.,Department of Translational Hematology and Oncology Research, Cleveland Clinic, OH, 44106, USA
| | - Jørn Skavland
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Baliakas Panagiotis
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Randi Hovland
- Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - Vibeke Andresen
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ole Morten Seternes
- Department of Pharmacy, UiT-The Arctic University of Norway, 9037, Tromsø, Norway
| | | | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, 94121, USA.,Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94121, USA.,Department of Biomedical Informatics, Stanford University School of Medicine, Stanford, CA, 94121, USA
| | - Sonia Gavasso
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Science, University of Bergen, Bergen, Norway.,Centre for Clinical Treatment Research (NeuroSysMed), Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Kimmo Porkka
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Inge Jonassen
- Centre for Cancer Biomarkers and Computational Biology Unit, Department of Informatics, University of Bergen, Bergen, Norway
| | - Yngvar Fløisand
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0318, Oslo, Norway.,Department of Hematology, Oslo University Hospital, Oslo, Norway
| | - Jorrit Enserink
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Montebello, 0379, Oslo, Norway.,Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0318, Oslo, Norway.,Section for Biochemistry and Molecular Biology, Faculty of Mathematics and Natural Sciences, University of Oslo, 0037, Oslo, Norway
| | - Nello Blaser
- Department of Informatics, University of Bergen, Bergen, Norway.
| | - Bjørn Tore Gjertsen
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Science, University of Bergen, Bergen, Norway. .,Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Helse Bergen HF, Bergen, Norway.
| |
Collapse
|
3
|
Singh A, Bhatia P, Trehan A, Bansal D, Singh A, Bhatia A. Low spontaneous apoptosis index at diagnosis predicts a high-risk phenotype in paediatric acute lymphoblastic leukaemia. Indian J Med Res 2018; 147:248-255. [PMID: 29923513 PMCID: PMC6022376 DOI: 10.4103/ijmr.ijmr_303_16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background & objectives: Significance of apoptosis as a prognostic marker is less well studied in paediatric acute lymphoblastic leukaemia (ALL) cases. Hence, a prospective study, involving 30 paediatric ALL cases, was done to assess the clinical relevance of in vivo apoptosis. Methods: Peripheral blood mononuclear cells from all patients were subjected to annexin V/propidium iodide staining to detect the degree of apoptosis [apoptotic index (AI)] at day 0 and day 35 post-induction chemotherapy. In addition, Bax and Bcl2 apoptotic protein expressions were studied at day 0 and their relative fluorescence mean intensity (RFMI) ratios were calculated. Results: Mean age of patients was 5.1 years. Of the 30 cases, 21 (70%) were at standard-risk, five (17%) at intermediate and four (13%) at high risk. Majority (83%) were B-ALL. Day 8 absolute blast count was >1000/μl in seven (23%) and <1000/μl in 23 of 30 (77%) cases. Day 35 marrow was M1 in 23 (92%) and M2 in two of 25 (8%) cases. AI at day 0 and day 35 ranged from 0.9 to16.6 per cent and 1.4 to 62.8 per cent with a mean of 5.90 and 19.64 per cent, respectively. The Bax/Bcl2 ratio ranged from 0.2 to 3.5 with a mean of 0.83. The ratio was predominantly anti-apoptotic, i.e. <1 (77%). A significant association was noted between low AI at day 0 and high total leucocyte count (P=0.02), T-cell phenotype (P=0.043) and high-risk as per NCI category (P=0.025). Significant increase (>30%) in day 35 AI was seen in only six cases. Interpretation & conclusions: Our study showed that low AI at day 0 was associated with a high-risk clinical phenotype in paediatric ALL. However, studies on larger group, especially with longer follow up or study of relapse cases, will help draw conclusions regarding apoptosis assessment in paediatric ALL.
Collapse
Affiliation(s)
- Ankita Singh
- Department of Paediatrics, Pediatric Hemato-Oncology Unit, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Prateek Bhatia
- Department of Paediatrics, Pediatric Hemato-Oncology Unit, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Amita Trehan
- Department of Paediatrics, Pediatric Hemato-Oncology Unit, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Deepak Bansal
- Department of Paediatrics, Pediatric Hemato-Oncology Unit, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Ajit Singh
- Department of Experimental Medicine, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Alka Bhatia
- Department of Experimental Medicine, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| |
Collapse
|
4
|
Song J, Seo Y, Park H. Pinosylvin enhances leukemia cell death via down-regulation of AMPKα expression. Phytother Res 2018; 32:2097-2104. [PMID: 30027566 DOI: 10.1002/ptr.6156] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 05/30/2018] [Accepted: 06/26/2018] [Indexed: 11/12/2022]
Abstract
Resveratrol at high concentrations (50-100 μmol/L) is known to induce cell death in leukemia cells. Here, we investigated whether pinosylvin, a resveratrol analogue, induced cell death in leukemia cells. Cell death was found to be markedly elevated by 50- to 100-μmol/L pinosylvin in THP-1 and U937 cells. It was also shown that pinosylvin induced caspase-3 activation, flip-flop of phosphatidylserine, LC3-II accumulation, LC3 puncta, and p62 degradation in both THP-1 and U937 cells. These data indicate that pinosylvin-induced cell death may occur through apoptosis and autophagy. In addition, we showed that pinosylvin down-regulates AMP-activated protein kinase α1 (AMPKα1) in leukemia cells. Therefore, we correlated AMPKα1 down-regulation and leukemia cell death. AMPKα1 inhibition appeared to decrease pinosylvin-induced apoptosis and autophagy in leukemia cells, implying that AMPK is a key regulator of leukemia cell death. Moreover, we found that both pinosylvin-induced autophagy and apoptotic progress were reduced in AMPKα1-overexpressed leukemia cells, when compared with vector-transfected cells. Cell death was elevated by AMPKα1 overexpression, whereas pinosylvin-induced cell death was markedly decreased by caspase-3 inhibitors or autophagy inhibitors. These results suggest that pinosylvin-induced depletion of AMPKα1 enhances cell death via apoptosis and autophagy in leukemia cells.
Collapse
Affiliation(s)
- Jina Song
- Department of Molecular Biology, Institute of Nanosensor and Biotechnology, Dankook University, Chungnam, South Korea
| | - Youngsik Seo
- Department of Molecular Biology, Institute of Nanosensor and Biotechnology, Dankook University, Chungnam, South Korea
| | - Heonyong Park
- Department of Molecular Biology, Institute of Nanosensor and Biotechnology, Dankook University, Chungnam, South Korea
| |
Collapse
|
5
|
Cabrera M, Gomez N, Remes Lenicov F, Echeverría E, Shayo C, Moglioni A, Fernández N, Davio C. G2/M Cell Cycle Arrest and Tumor Selective Apoptosis of Acute Leukemia Cells by a Promising Benzophenone Thiosemicarbazone Compound. PLoS One 2015; 10:e0136878. [PMID: 26360247 PMCID: PMC4567328 DOI: 10.1371/journal.pone.0136878] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 08/09/2015] [Indexed: 01/10/2023] Open
Abstract
Anti-mitotic therapies have been considered a hallmark in strategies against abnormally proliferating cells. Focusing on the extensively studied family of thiosemicarbazone (TSC) compounds, we have previously identified 4,4'-dimethoxybenzophenone thiosemicarbazone (T44Bf) as a promising pharmacological compound in a panel of human leukemia cell lines (HL60, U937, KG1a and Jurkat). Present findings indicate that T44Bf-mediated antiproliferative effects are associated with a reversible chronic mitotic arrest caused by defects in chromosome alignment, followed by induced programmed cell death. Furthermore, T44Bf selectively induces apoptosis in leukemia cell lines when compared to normal peripheral blood mononuclear cells. The underlying mechanism of action involves the activation of the mitochondria signaling pathway, with loss of mitochondrial membrane potential and sustained phosphorylation of anti-apoptotic protein Bcl-xL as well as increased Bcl-2 (enhanced phosphorylated fraction) and pro-apoptotic protein Bad levels. In addition, ERK signaling pathway activation was found to be a requisite for T44Bf apoptotic activity. Our findings further describe a novel activity for a benzophenone thiosemicarbazone and propose T44Bf as a promising anti-mitotic prototype to develop chemotherapeutic agents to treat acute leukemia malignancies.
Collapse
Affiliation(s)
- Maia Cabrera
- Instituto de Investigaciones Farmacológicas, Facultad de Farmacia y Bioquímica (ININFA-UBA-CONICET), Buenos Aires, Argentina
- * E-mail:
| | - Natalia Gomez
- Instituto de Investigaciones Farmacológicas, Facultad de Farmacia y Bioquímica (ININFA-UBA-CONICET), Buenos Aires, Argentina
- Cátedra de Química Medicinal, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Federico Remes Lenicov
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA, Facultad de Medicina, (INBIRS-UBA-CONICET), Buenos Aires, Argentina
| | - Emiliana Echeverría
- Instituto de Investigaciones Farmacológicas, Facultad de Farmacia y Bioquímica (ININFA-UBA-CONICET), Buenos Aires, Argentina
| | - Carina Shayo
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Albertina Moglioni
- Instituto de Química y Metabolismo del Fármaco, Facultad de Farmacia y Bioquímica, (IQUIMEFA-UBA-CONICET), Buenos Aires, Argentina
- Cátedra de Química Medicinal, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Natalia Fernández
- Instituto de Investigaciones Farmacológicas, Facultad de Farmacia y Bioquímica (ININFA-UBA-CONICET), Buenos Aires, Argentina
- Cátedra de Química Medicinal, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carlos Davio
- Instituto de Investigaciones Farmacológicas, Facultad de Farmacia y Bioquímica (ININFA-UBA-CONICET), Buenos Aires, Argentina
- Cátedra de Química Medicinal, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
6
|
Boppana NB, Stochaj U, Kodiha M, Bielawska A, Bielawski J, Pierce JS, Korbelik M, Separovic D. Enhanced killing of SCC17B human head and neck squamous cell carcinoma cells after photodynamic therapy plus fenretinide via the de novo sphingolipid biosynthesis pathway and apoptosis. Int J Oncol 2015; 46:2003-10. [PMID: 25739041 PMCID: PMC4383026 DOI: 10.3892/ijo.2015.2909] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 01/05/2015] [Indexed: 02/05/2023] Open
Abstract
Because photodynamic therapy (PDT) alone is not always effective as an anticancer treatment, PDT is combined with other anticancer agents for improved efficacy. The clinically-relevant fenretinide [N-(4-hydroxyphenyl) retinamide; 4HPR], was combined with the silicon phthalocyanine photosensitizer Pc4-mediated PDT to test for their potential to enhance killing of SCC17B cells, a clinically-relevant model of human head and neck squamous cell carcinoma. Because each of these treatments induces apoptosis and regulates the de novo sphingolipid (SL) biosynthesis pathway, the role of ceramide synthase, the pathway-associated enzyme, in PDT+4HPR-induced apoptotic cell death was determined using the ceramide synthase inhibitor fumonisin B1 (FB). PDT+4HPR enhanced loss of clonogenicity. zVAD-fmk, a pan-caspase inhibitor, and FB, protected cells from death post-PDT+4HPR. In contrast, the anti-apoptotic protein Bcl2 inhibitor ABT199 enhanced cell killing after PDT+4HPR. Combining PDT with 4HPR led to FB-sensitive, enhanced Bax associated with mitochondria and cytochrome c redistribution. Mass spectrometry data showed that the accumulation of C16-dihydroceramide, a precursor of ceramide in the de novo SL biosynthesis pathway, was enhanced after PDT+4HPR. Using quantitative confocal microscopy, we found that PDT+4HPR enhanced dihydroceramide/ceramide accumulation in the ER, which was inhibited by FB. The results suggest that SCC17B cells are sensitized to PDT by 4HPR via the de novo SL biosynthesis pathway and apoptosis, and imply potential clinical relevance of the combination for cancer treatment.
Collapse
Affiliation(s)
- Nithin B Boppana
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Ursula Stochaj
- Department of Physiology, McGill University, Montreal, QC H3G 1YC, Canada
| | - Mohamed Kodiha
- Department of Physiology, McGill University, Montreal, QC H3G 1YC, Canada
| | - Alicja Bielawska
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Jacek Bielawski
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Jason S Pierce
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Mladen Korbelik
- British Columbia Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Duska Separovic
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
7
|
Del Principe MI, Del Poeta G, Venditti A, Buccisano F, Maurillo L, Mazzone C, Bruno A, Neri B, Irno Consalvo M, Lo Coco F, Amadori S. Apoptosis and immaturity in acute myeloid leukemia. Hematology 2013; 10:25-34. [PMID: 16019442 DOI: 10.1080/10245330400020454] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
The primary cause of treatment failures in acute myeloid leukemia (AML) is the emergence of both resistant disease and early relapse. Among the most frequent agents of these phenomena are defects in the mitochondrial-mediated apoptotic pathway. This pathway is regulated by bcl-2 family of anti-apoptotic (bcl-2, bcl-xl, mcl-1) and pro-apoptotic proteins (bax, bad, bak). In particular, bcl-2 dimerizes with several members of bcl-2 family of proteins, altering the threshold of cell death. The flow cytometric quantitative measurement of bcl-2 and bax expression for the determination of bax/bcl-2 ratio provided crucial clinical information in AML: in our hands, lower bax/bcl-2 ratio conferred a very poor prognosis with decreased rates of complete remission (CR) and overall survival (OS). Moreover, striking correlations were found between lower bax/bcl-2 ratio and higher progenitor marker expression, such as CD34, CD117 and CD133 antigens, confirming the link between this apoptotic index and the maturation pathways. However, the capacity of bax/bcl-2 ratio to clearly identify patients with different prognosis with regard to CR and OS within the CD34+, CD117+ and CD133+ subgroups implies that other mechanisms, such as proliferation and/or cell cycle dysregulation may be involved to explain its clinical significance. Finally, small molecules that target both the receptor- and mitochondrial-mediated pathway of apoptosis are providing encouraging results in patients with relapsed and/or refractory disease (i.e. CDDOMe, bcl-2 antisense oligonucleotides, CEP-701, etc), confirming the key role of apoptotic mechanisms on the outcome of AML patients.
Collapse
|
8
|
Chueh FS, Hsiao YT, Chang SJ, Wu PP, Yang JS, Lin JJ, Chung JG, Lai TY. Glycyrrhizic acid induces apoptosis in WEHI-3 mouse leukemia cells through the caspase- and mitochondria-dependent pathways. Oncol Rep 2012; 28:2069-76. [PMID: 22972479 DOI: 10.3892/or.2012.2029] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 08/07/2012] [Indexed: 11/06/2022] Open
Abstract
Leukemia, one of the causes of cancer-related death in humans, is an aggressive malignancy via the rapid growth of abnormal white blood cells. The aim of this study was to determine the anti-leukemia effect of glycyrrhizic acid (GA) on a mouse leukemia cell line, WEHI-3. GA, an active compound in Glycyrrhiza glabra, has been proven to induce cytotoxic effects in many cancer cell lines. In the current study, we investigated the effects of GA in mouse leukemia cells in vitro. The results indicated that GA induced morphological changes, G0/G1 phase arrest, apoptosis and DNA damage in WEHI-3 cells as determined by phase contrast microscopy, DAPI-staining, flow cytometry and comet assay. The results from the flow cytometric assay showed that GA increased ROS levels, reduced the mitochondrial membrane potential (ΔΨm) and stimulated caspase-3 activity in WEHI-3 cells. GA regulated the intrinsic and extrinsic apoptosis-associated protein expression which was determined by western blotting. In addition, endoplasmic reticulum (ER) stress responses were observed in GA-treated WEHI-3 cells. GA promoted the trafficking of apoptosis-inducing factor (AIF), cytochrome c and endonuclease G (Endo G) in WEHI-3 cells. Based on this evidence, GA-triggered apoptosis occurs through the death receptor, mitochondria-mediated and ER stress multiple signaling pathways.
Collapse
Affiliation(s)
- Fu-Shin Chueh
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan, ROC
| | | | | | | | | | | | | | | |
Collapse
|
9
|
LEE CHAOYING, CHIEN YUNGSHIN, CHIU TAIHUI, HUANG WENWEN, LU CHICHENG, CHIANG JOHUA, YANG JAISING. Apoptosis triggered by vitexin in U937 human leukemia cells via a mitochondrial signaling pathway. Oncol Rep 2012; 28:1883-8. [DOI: 10.3892/or.2012.2000] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 07/24/2012] [Indexed: 11/06/2022] Open
|
10
|
Lu CC, Yang JS, Chiang JH, Hour MJ, Lin KL, Lin JJ, Huang WW, Tsuzuki M, Lee TH, Chung JG. Novel quinazolinone MJ-29 triggers endoplasmic reticulum stress and intrinsic apoptosis in murine leukemia WEHI-3 cells and inhibits leukemic mice. PLoS One 2012; 7:e36831. [PMID: 22662126 PMCID: PMC3360742 DOI: 10.1371/journal.pone.0036831] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Accepted: 04/07/2012] [Indexed: 01/11/2023] Open
Abstract
The present study was to explore the biological responses of the newly compound, MJ-29 in murine myelomonocytic leukemia WEHI-3 cells in vitro and in vivo fates. We focused on the in vitro effects of MJ-29 on ER stress and mitochondria-dependent apoptotic death in WEHI-3 cells, and to hypothesize that MJ-29 might fully impair the orthotopic leukemic mice. Our results indicated that a concentration-dependent decrease of cell viability was shown in MJ-29-treated cells. DNA content was examined utilizing flow cytometry, whereas apoptotic populations were determined using annexin V/PI, DAPI staining and TUNEL assay. Increasing vital factors of mitochondrial dysfunction by MJ-29 were further investigated. Thus, MJ-29-provaked apoptosis of WEHI-3 cells is mediated through the intrinsic pathway. Importantly, intracellular Ca2+ release and ER stress-associated signaling also contributed to MJ-29-triggered cell apoptosis. We found that MJ-29 stimulated the protein levels of calpain 1, CHOP and p-eIF2α pathways in WEHI-3 cells. In in vivo experiments, intraperitoneal administration of MJ-29 significantly improved the total survival rate, enhanced body weight and attenuated enlarged spleen and liver tissues in leukemic mice. The infiltration of immature myeloblastic cells into splenic red pulp was reduced in MJ-29-treated leukemic mice. Moreover, MJ-29 increased the differentiations of T and B cells but decreased that of macrophages and monocytes. Additionally, MJ-29-stimulated immune responses might be involved in anti-leukemic activity in vivo. Based on these observations, MJ-29 suppresses WEHI-3 cells in vitro and in vivo, and it is proposed that this potent and selective agent could be a new chemotherapeutic candidate for anti-leukemia in the future.
Collapse
Affiliation(s)
- Chi-Cheng Lu
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Jai-Sing Yang
- Department of Pharmacology, China Medical University, Taichung, Taiwan
| | - Jo-Hua Chiang
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Mann-Jen Hour
- School of Pharmacy, China Medical University, Taichung, Taiwan
| | - Kuei-Li Lin
- Department of Radiation Oncology, Chi Mei Medical Center, Tainan, Taiwan
| | - Jen-Jyh Lin
- Graduate Institute of Chinese Medicine, China Medical University, Taichung, Taiwan
- Division of Cardiology, China Medical University Hospital, Taichung, Taiwan
| | - Wen-Wen Huang
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Minoru Tsuzuki
- Department of Biochemistry, Nihon Pharmaceutical University, Saitama, Japan
- Tsuzuki Institute for Traditional Medicine, China Medical University, Taichung, Taiwan
| | - Tsung-Han Lee
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
- * E-mail: (JGC); (THL)
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
- * E-mail: (JGC); (THL)
| |
Collapse
|
11
|
Shigemizu D, Hu Z, Hung JH, Huang CL, Wang Y, DeLisi C. Using functional signatures to identify repositioned drugs for breast, myelogenous leukemia and prostate cancer. PLoS Comput Biol 2012; 8:e1002347. [PMID: 22346740 PMCID: PMC3276504 DOI: 10.1371/journal.pcbi.1002347] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Accepted: 11/25/2011] [Indexed: 01/21/2023] Open
Abstract
The cost and time to develop a drug continues to be a major barrier to widespread distribution of medication. Although the genomic revolution appears to have had little impact on this problem, and might even have exacerbated it because of the flood of additional and usually ineffective leads, the emergence of high throughput resources promises the possibility of rapid, reliable and systematic identification of approved drugs for originally unintended uses. In this paper we develop and apply a method for identifying such repositioned drug candidates against breast cancer, myelogenous leukemia and prostate cancer by looking for inverse correlations between the most perturbed gene expression levels in human cancer tissue and the most perturbed expression levels induced by bioactive compounds. The method uses variable gene signatures to identify bioactive compounds that modulate a given disease. This is in contrast to previous methods that use small and fixed signatures. This strategy is based on the observation that diseases stem from failed/modified cellular functions, irrespective of the particular genes that contribute to the function, i.e., this strategy targets the functional signatures for a given cancer. This function-based strategy broadens the search space for the effective drugs with an impressive hit rate. Among the 79, 94 and 88 candidate drugs for breast cancer, myelogenous leukemia and prostate cancer, 32%, 13% and 17% respectively are either FDA-approved/in-clinical-trial drugs, or drugs with suggestive literature evidences, with an FDR of 0.01. These findings indicate that the method presented here could lead to a substantial increase in efficiency in drug discovery and development, and has potential application for the personalized medicine. The effective drug of a given disease is aimed to bring abnormal functions associated with disease back to the normal state. Using expression profile as the surrogate marker of the cellular function, we introduce a novel procedure to identify candidate therapeutics by searching for those bioactive compounds that either down-regulate abnormally over-expressed genes, or up-regulate those that are abnormally under-expressed. We show that the approach detects a pool of plausible candidates as repositioning/new drugs. In contrast to previous studies, our approach uses a variable big number of genes and/or gene combinations as a representation of functional signatures to identify bioactive compounds that modulate a given disease, irrespective of the particular genes that contribute to the cellular functions; therefore it covers potential drugs with heterogeneous properties. The method may also have potential application for the personalized medicine.
Collapse
Affiliation(s)
- Daichi Shigemizu
- Bioinformatics Program, Boston University, Boston, Massachusetts, United States of America
| | - Zhenjun Hu
- Bioinformatics Program, Boston University, Boston, Massachusetts, United States of America
| | - Jui-Hung Hung
- Bioinformatics Program, Boston University, Boston, Massachusetts, United States of America
| | - Chia-Ling Huang
- Bioinformatics Program, Boston University, Boston, Massachusetts, United States of America
| | - Yajie Wang
- Laboratory of Clinical Medical Research, Department of Clinical Laboratory Diagnostics, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Charles DeLisi
- Bioinformatics Program, Boston University, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
12
|
Øbro NF, Ryder LP, Madsen HO, Andersen MK, Lausen B, Hasle H, Schmiegelow K, Marquart HV. Identification of residual leukemic cells by flow cytometry in childhood B-cell precursor acute lymphoblastic leukemia: verification of leukemic state by flow-sorting and molecular/cytogenetic methods. Haematologica 2011; 97:137-41. [PMID: 21933850 DOI: 10.3324/haematol.2011.051383] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Reduction in minimal residual disease, measured by real-time quantitative PCR or flow cytometry, predicts prognosis in childhood B-cell precursor acute lymphoblastic leukemia. We explored whether cells reported as minimal residual disease by flow cytometry represent the malignant clone harboring clone-specific genomic markers (53 follow-up bone marrow samples from 28 children with B-cell precursor acute lymphoblastic leukemia). Cell populations (presumed leukemic and non-leukemic) were flow-sorted during standard flow cytometry-based minimal residual disease monitoring and explored by PCR and/or fluorescence in situ hybridization. We found good concordance between flow cytometry and genomic analyses in the individual flow-sorted leukemic (93% true positive) and normal (93% true negative) cell populations. Four cases with discrepant results had plausible explanations (e.g. partly informative immunophenotype and antigen modulation) that highlight important methodological pitfalls. These findings demonstrate that with sufficient experience, flow cytometry is reliable for minimal residual disease monitoring in B-cell precursor acute lymphoblastic leukemia, although rare cases require supplementary PCR-based monitoring.
Collapse
Affiliation(s)
- Nina F Øbro
- Department of Clinical Immunology, University Hospital Rigshospitalet, Copenhagen, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Immunophenotype-defined sub-populations are common at diagnosis in childhood B-cell precursor acute lymphoblastic leukemia. Leukemia 2011; 25:1652-7. [DOI: 10.1038/leu.2011.136] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
14
|
Synthesis, Structural Characterization, and Pro-apoptotic Activity of 1-Indanone Thiosemicarbazone Platinum(II) and Palladium(II) Complexes: Potential as Antileukemic Agents. ChemMedChem 2011; 6:1485-94. [DOI: 10.1002/cmdc.201100060] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Revised: 04/26/2011] [Indexed: 11/07/2022]
|
15
|
Monitoring of T-cell acute lymphoblastic leukemia by flow cytometry. Open Med (Wars) 2010. [DOI: 10.2478/s11536-010-0044-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AbstractMinimal residual disease (MRD) predicts the outcome of acute lymphoblastic leukemia (ALL). Flow cytometry (FC) is one of the most sensitive and most applicable methods for MRD diagnostics, but there is still no agreement on the “gold standard” of the method. We tried to optimize flow cytometric MRD detection in T-ALL. Fourteen adults and 11 children with T-ALL and 12 normal bone marrow (BM) donors were enrolled in the study. We found that the most common phenotypic aberrations in T-ALL were TdT and CD99 coexpression on T-cells in BM. Therefore for MRD detection we developed a limited four-color marker panel (TdT/CD7/cCD3/CD19 and CD99/CD7/cCD3/CD2) and a standard analysis strategy. This assay was evaluated on BM of healthy controls. Less than 0.01% TdT+ or CD99 bright T-cells were found in normal BM. MRD was detected in 9 adult patients and 1 child at different time-points of treatment. The average TdT and CD99 mean fluorescence intensity (MFI) value of residual blasts fluctuated during therapy, but it still remained higher than MFI of normal T-cells. Our established MRD detection method differentiated leukemic lymphoblasts with sensitivity in the range of 0.01% and did not give any false positive results in normal BM.
Collapse
|
16
|
Dworzak MN, Gaipa G, Schumich A, Maglia O, Ratei R, Veltroni M, Husak Z, Basso G, Karawajew L, Gadner H, Biondi A. Modulation of antigen expression in B-cell precursor acute lymphoblastic leukemia during induction therapy is partly transient: evidence for a drug-induced regulatory phenomenon. Results of the AIEOP-BFM-ALL-FLOW-MRD-Study Group. CYTOMETRY PART B-CLINICAL CYTOMETRY 2010; 78:147-53. [PMID: 20201055 DOI: 10.1002/cyto.b.20516] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Changes of antigen expression on residual blast cells of acute lymphoblastic leukemia (ALL) occur during induction treatment. Many markers used for phenotyping and minimal residual disease (MRD) monitoring are affected. Glucocorticoid (GC)-induced expression modulation has been causally suspected, however, subclone selection may also cause the phenomenon. METHODS We investigated this by following the phenotypic evolution of leukemic cells with flow cytometry from diagnosis to four time points during and after GC containing chemotherapy in the 20 (of 360 consecutive) B-cell precursor patients with ALL who had persistent MRD throughout. RESULTS The early expression changes of CD10 and CD34 were reversible after stop of GC containing chemotherapy. Modulation of CD20 and CD45 occurred mostly during the GC phase, whereas CD11a also changed later on. Blast cells at diagnosis falling into gates designed according to "shifted" phenotypes from follow-up did not form clusters and were frequently less numerous than later on. CONCLUSIONS Our data support the idea that drug-induced modulation rather than selection causes the phenomenon. The good message for MRD assessment is that modulation is transient in at least two (CD10 and CD34) of the five prominent antigens investigated and reverts to initial aberrant patterns after stop of GC therapy, whereas CD20 expression gains new aberrations exploitable for MRD detection.
Collapse
Affiliation(s)
- Michael N Dworzak
- Laboratory of Immunological Diagnostics, Children's Cancer Research Institute, Vienna, Austria.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
CD11b is a therapy resistance- and minimal residual disease-specific marker in precursor B-cell acute lymphoblastic leukemia. Blood 2010; 115:3763-71. [PMID: 20228269 DOI: 10.1182/blood-2009-10-247585] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
A consistently increased mRNA expression of the adhesion receptor CD11b is a hallmark of the reported genomewide gene expression changes in precursor B-cell acute lymphoblastic leukemia (PBC-ALL) after 1 week of induction therapy. To investigate its clinical relevance, CD11b protein expression in leukemic blasts has been prospectively measured at diagnosis (159 patients) and during therapy (53 patients). The initially heterogeneous expression of CD11b inversely correlated with cytoreduction rates measured at clinically significant time points of induction therapy in the ALL-Berlin-Frankfurt-Münster 2000 protocol. CD11b positivity conferred a 5-fold increased risk of minimal residual disease (MRD) after induction therapy (day 33) and of high-risk group assignment after consolidation therapy (day 78). In the multivariate analysis CD11b expression was an independent prognostic factor compared with other clinically relevant parameters at diagnosis. During therapy, CD11b expression increased early in most ALL cases and remained consistently increased during induction/consolidation therapy. In more than 30% of MRD-positive cases, the CD11b expression on blast cells exceeded that of mature memory B cells and improved the discrimination of residual leukemic cells from regenerating bone marrow. Taken together, CD11b expression has considerable implications for prognosis, treatment response monitoring, and MRD detection in childhood PBC-ALL.
Collapse
|
18
|
Dakka N, Bellaoui H, Bouzid N, Khattab M, Bakri Y, Benjouad A. CD10 AND CD34 expression in childhood acute lymphoblastic leukemia in Morocco: clinical relevance and outcome. Pediatr Hematol Oncol 2009; 26:216-31. [PMID: 19437324 DOI: 10.1080/07357900902897557] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
CD10 and CD34 expression in 86 Moroccan children with acute lymphoblastic leukemias (ALL) and the relevance to prognosis, diagnosis, and outcome during a 5-year follow-up were examined. At diagnosis, 57% of patients had CD10(+) blasts, while 35% had CD34(+) blasts. The CD10(+) blast frequency was much higher (80%) in B-ALL than in T-ALL (20%). The frequency of CD34(+) blasts was higher in B-ALL (48%) compared to T-ALL (16%). The 5-year survival curves showed that children with CD10(+) B-ALL had a significantly longer survival rate than those with CD10(-), as observed for T-ALL. The survival rate of B-ALL expressing CD34 was higher than that of CD34(-). Thus, CD34 and CD10 expression may have prognostic value and is associated with a better clinical outcome.
Collapse
Affiliation(s)
- Nadia Dakka
- Laboratoire de Biochimie et Immunologie, Faculté des Sciences, Université Mohammed V, Rabat, Morocco.
| | | | | | | | | | | |
Collapse
|
19
|
CD34+ leukemic subpopulation predominantly displays lower spontaneous apoptosis and has higher expression levels of Bcl-2 and MDR1 genes than CD34- cells in childhood AML. Ann Hematol 2008; 87:353-60. [PMID: 18228020 DOI: 10.1007/s00277-008-0439-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2007] [Accepted: 01/02/2008] [Indexed: 10/22/2022]
Abstract
In view of obscure clinical and biological significance of leukemic cells heterogeneity, we studied the efficacy of apoptosis, proliferation, and expression levels of the Bcl-2, MDR1, LRP, and BCRP genes in sorted CD34+ and CD34- subpopulations of childhood AML leukemic samples. In five out of nine cases, CD34+ cells were less sensitive to spontaneous apoptosis and had from 1.2- to 5.0-fold higher expression levels of Bcl-2 (eight of ten) and from 1.5- to 28.7-fold higher expression levels of MDR1 (eight of ten). The expression levels of the LRP gene were from 1.1- to 1.8-fold higher in CD34+ subpopulations (five of ten cases), and the expression levels of the BCRP gene were from 1.1- to 22.4-fold higher in CD34+ leukemic cells (six of ten). In all M4 cases, the expression levels of LRP were higher in the CD34- subpopulation. Significant differences in the patterns of genes expression between patients do not allow us to conclude that the CD34+ fractions have more resistant phenotype than the CD34- subpopulations. Nevertheless, distinctions between CD34+ and CD34- cells may lead to different chemosensitivities between leukemic subpopulations in vivo and may determine the alteration of the leukemic immunophenotype during treatment and in relapse.
Collapse
|
20
|
Mueller S, Holdenrieder S, Stieber P, Haferlach T, Schalhorn A, Braess J, Nagel D, Seidel D. Early prediction of therapy response in patients with acute myeloid leukemia by nucleosomal DNA fragments. BMC Cancer 2006; 6:143. [PMID: 16734907 PMCID: PMC1555596 DOI: 10.1186/1471-2407-6-143] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2005] [Accepted: 05/30/2006] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Elevated levels of nucleosomal DNA fragments can be detected in plasma and sera of patients with malignant diseases. METHODS We investigated the course of nucleosomal DNA, thymidine kinase, lactate dehydrogenase and leukocytes in sera of 25 patients with acute myeloid leukemia during the first cycle of induction chemotherapy and tested their power to distinguish between patients with complete remission and those with no remission. RESULTS Almost all patients showed strongly decreasing levels of nucleosomal DNA during the first week, in some cases after initial peaks. In overall analysis of variance, DNA levels could clearly distinguish between patients with complete remission, who had higher DNA values, and those with insufficient response (p = 0.017). The area under the curve of DNA values of days 2-4 after start of therapy (AUC 2-4) discriminated between both groups with a sensitivity of 56% at a specificity of 100%. Further, pretherapeutic levels and AUC 2-4 of nucleosomal DNA correlated significantly with blast reduction after 16 days. A tendency to higher levels in patients with complete response was also found for thymidine kinase, lactate dehydrogenase and leukocytes, however the difference did not reach the level of significance (p = 0.542, p = 0.260, and p = 0.144, respectively). CONCLUSION Our results indicate that nucleosomal DNA fragments are valuable markers for the early prediction of therapeutic efficacy in patients with acute myeloid leukemia.
Collapse
Affiliation(s)
- Susanne Mueller
- Institute of Clinical Chemistry, University Hospital Munich-Grosshadern, Marchioninistr. 15, D-81377 Munich, Germany
| | - Stefan Holdenrieder
- Institute of Clinical Chemistry, University Hospital Munich-Grosshadern, Marchioninistr. 15, D-81377 Munich, Germany
| | - Petra Stieber
- Institute of Clinical Chemistry, University Hospital Munich-Grosshadern, Marchioninistr. 15, D-81377 Munich, Germany
| | - Torsten Haferlach
- Department of Internal Medicine III, University Hospital Munich-Grosshadern, Marchioninistr. 15, D-81377 Munich, Germany
| | - Andreas Schalhorn
- Department of Internal Medicine III, University Hospital Munich-Grosshadern, Marchioninistr. 15, D-81377 Munich, Germany
| | - Jan Braess
- Department of Internal Medicine III, University Hospital Munich-Grosshadern, Marchioninistr. 15, D-81377 Munich, Germany
| | - Dorothea Nagel
- Institute of Clinical Chemistry, University Hospital Munich-Grosshadern, Marchioninistr. 15, D-81377 Munich, Germany
| | - Dietrich Seidel
- Institute of Clinical Chemistry, University Hospital Munich-Grosshadern, Marchioninistr. 15, D-81377 Munich, Germany
| |
Collapse
|
21
|
Vial JP, Tabrizi R, Pigneux A, Lacombe F, Praloran V, Belloc F. Remission induction chemotherapy induces in vivo caspase-dependent apoptosis in bone marrow acute myeloid leukemia blast cells and spares lymphocytes. CYTOMETRY PART B-CLINICAL CYTOMETRY 2006; 70:115-23. [PMID: 16572429 DOI: 10.1002/cyto.b.20105] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND The goal of new therapeutic strategies is to adapt the treatment of acute myeloid leukemia (AML) patients to the prognostic and/or to the hematological response. METHODS We analyzed in vivo apoptosis induction in blast cells and in lymphocytes of AML patients receiving remission induction treatment. RESULTS We show, on 12 peripheral blood samples, that the increase of peripheral apoptotic blast cells cannot be considered as the earliest marker of the treatment efficiency, because the significant increase of apoptosis followed the white blood cell and the peripheral blast cell count reductions, probably due to an efficient clearance of circulating apoptotic cells. Furthermore, the study of 65 bone marrow samples at d15 showed that the treatment induced apoptosis of blast cells while sparing the lymphocytes. This apoptosis was evidenced both at the caspase and at the membrane levels using respectively fmk-VAD-FITC and Annexin V binding assays. We found that less than 50% of apoptosis, measured with the fmk-VAD-FITC, in the d15 residual bone marrow blast cells, correlated with lower disease-free survival probability. CONCLUSION More studies are needed in larger series and earlier during the remission induction treatment to confirm the possible prognostic significance of in vivo apoptosis induction.
Collapse
Affiliation(s)
- J-P Vial
- Laboratoire d'Hématologie, CHU de Bordeaux Hôpital du Haut-Lévêque, Pessac, France.
| | | | | | | | | | | |
Collapse
|
22
|
Kim HJ, Park BH, Choi Y, Min WS, Lee JW, Kim CC. Fas, Fas-Associated Death Domain-Like Interleukin 1β -Converting Enzyme-Like Inhibitory Protein, and Apoptotic Features of Elderly Acute Myeloid Leukemia Based on Response to Induction Chemotherapy. Int J Hematol 2005; 82:327-32. [PMID: 16298825 DOI: 10.1532/ijh97.a30503] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
A study was performed to examine the clinical outcome of triple-combination induction chemotherapy in 26 elderly Korean acute myeloid leukemia (AML) patients and to investigate apoptotic responses during and after treatment to determine whether the responses can be used as prognostic markers. Patients who had Western blot or polymerase chain reaction analysis findings of higher expression levels of Fas-associated death domain-like interleukin 1beta-converting enzyme-like inhibitory protein (FLIP) on day 7 after chemotherapy were more likely to have complete remission, but there was less or no correlation with Fas or a proapoptosis/apoptosis reaction. Expression of FLIP molecules may be, at least in part, an early prognostic indicator in the treatment of elderly AML patients.
Collapse
MESH Headings
- Aged
- Antineoplastic Agents/therapeutic use
- Apoptosis
- Bone Marrow Cells/pathology
- CASP8 and FADD-Like Apoptosis Regulating Protein
- Enzyme Inhibitors/analysis
- Gene Expression Regulation, Neoplastic
- Humans
- In Situ Hybridization, Fluorescence
- Intracellular Signaling Peptides and Proteins/analysis
- Intracellular Signaling Peptides and Proteins/genetics
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/pathology
- Polymerase Chain Reaction
- Probability
- RNA, Messenger/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Survival Analysis
- Survivors
- fas Receptor/analysis
Collapse
Affiliation(s)
- Hee-Je Kim
- Division of Hematology, Department of Internal Medicine, Catholic Hemopoietic Stem Cell Transplantation Center, Catholic University of Korea College of Medicine, Seoul
| | | | | | | | | | | |
Collapse
|
23
|
Díaz LF, Chiong M, Quest AFG, Lavandero S, Stutzin A. Mechanisms of cell death: molecular insights and therapeutic perspectives. Cell Death Differ 2005; 12:1449-56. [PMID: 16052234 DOI: 10.1038/sj.cdd.4401738] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- L F Díaz
- Centro FONDAP de Estudios Moleculares de la Célula, Facultades de Medicina y Ciencias Químicas y Farmacéuticas, Universidad de Chile, Independencia 1027, Santiago 838-0453, Chile
| | | | | | | | | |
Collapse
|
24
|
Fulda S, Debatin KM. Sensitization for anticancer drug-induced apoptosis by betulinic Acid. Neoplasia 2005; 7:162-70. [PMID: 15802021 PMCID: PMC1501129 DOI: 10.1593/neo.04442] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We previously described that betulinic acid (BetA), a naturally occurring pentacyclic triterpenoid, induces apoptosis in tumor cells through the mitochondrial pathway. Here, for the first time, we provide evidence that BetA cooperated with anticancer drugs to induce apoptosis and to inhibit clonogenic survival of tumor cells. Combined treatment with BetA and anticancer drugs acted in concert to induce loss of mitochondrial membrane potential and the release of cytochrome c and Smac from mitochondria, resulting in activation of caspases and apoptosis. Overexpression of Bcl-2, which blocked mitochondrial perturbations, also inhibited the cooperative effect of BetA and anticancer drugs, indicating that cooperative interaction involved the mitochondrial pathway. Notably, cooperation of BetA and anticancer drugs was found for various cytotoxic compounds with different modes of action (e.g., doxorubicin, cisplatin, Taxol, VP16, or actino-mycin D). Importantly, BetA and anticancer drugs cooperated to induce apoptosis in different tumor cell lines, including p53 mutant cells, and also in primary tumor cells, but not in human fibroblasts indicating some tumor specificity. These findings indicate that using BetA as sensitizer in chemotherapy-based combination regimens may be a novel strategy to enhance the efficacy of anticancer therapy, which warrants further investigation.
Collapse
Affiliation(s)
- Simone Fulda
- University Children's Hospital, Prittwitzstrasse 43, Ulm 89075, Germany.
| | | |
Collapse
|
25
|
Schmidt S, Rainer J, Ploner C, Presul E, Riml S, Kofler R. Glucocorticoid-induced apoptosis and glucocorticoid resistance: molecular mechanisms and clinical relevance. Cell Death Differ 2005; 11 Suppl 1:S45-55. [PMID: 15243581 DOI: 10.1038/sj.cdd.4401456] [Citation(s) in RCA: 262] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The ability of glucocorticoids (GC) to efficiently kill lymphoid cells has led to their inclusion in essentially all chemotherapy protocols for lymphoid malignancies. This review summarizes recent findings related to the molecular basis of GC-induced apoptosis and GC resistance, and discusses their potential clinical implications. Accumulating evidence suggests that GC may induce cell death via different pathways resulting in apoptotic or necrotic morphologies, depending on the availability/responsiveness of the apoptotic machinery. The former might result from regulation of typical apoptosis genes such as members of the Bcl-2 family, the latter from detrimental GC effects on essential cellular functions possibly perpetuated by GC receptor (GR) autoinduction. Although other possibilities exist, GC resistance might frequently result from defective GR expression, perhaps the most efficient means to target multiple antileukemic GC effects. Numerous novel drug combinations are currently being tested to prevent resistance and improve GC efficacy in the therapy of lymphoid malignancies.
Collapse
Affiliation(s)
- S Schmidt
- Tyrolean Cancer Research Institute, Innsbruck, Austria
| | | | | | | | | | | |
Collapse
|
26
|
Gaipa G, Basso G, Maglia O, Leoni V, Faini A, Cazzaniga G, Bugarin C, Veltroni M, Michelotto B, Ratei R, Coliva T, Valsecchi MG, Biondi A, Dworzak MN. Drug-induced immunophenotypic modulation in childhood ALL: implications for minimal residual disease detection. Leukemia 2005; 19:49-56. [PMID: 15538405 DOI: 10.1038/sj.leu.2403559] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Assessment of minimal residual disease (MRD) by flow cytometry is considered to be based on the reproducibility of the leukemic immunophenotype detected at diagnosis. However, we previously noticed modulation of surface antigen expression in acute lymphoblastic leukemia (ALL) during the early treatment. Hence, we investigated this in 30 children with B-cell precursor ALL consecutively enrolled in the AIEOP-BFM ALL 2000 protocol. Quantitative expression of seven antigens useful in MRD monitoring was studied at diagnosis and compared to that measured at different time points of remission induction therapy. Downmodulation in the expression of CD10 and CD34 occurred at follow-up. By contrast, upmodulation of CD19, CD20, CD45RA, and CD11a was observed, while the expression of CD58 remained stable. Despite this, we could unambiguously discriminate leukemic cells from normal residual B cells. This holds true when bone marrow (BM) samples from similarly treated T-ALL patients, but not from healthy donors, were used as reference. Our results indicate that immunophenotypic modulation occurs in ALL during the early phases of BFM-type protocols. However, the accuracy of MRD detection by flow cytometry seems not negatively affected if adequate analysis protocols are employed. Investigators should take this phenomenon into account in order to avoid pitfalls in flow cytometric MRD studies.
Collapse
Affiliation(s)
- G Gaipa
- Centro Ricerca M Tettamanti, Clinica Pediatrica Università Milano-Bicocca, Ospedale San Gerardo, Via Donizetti 106, 20052 Monza, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Fulda S, Jeremias I, Debatin KM. Cooperation of betulinic acid and TRAIL to induce apoptosis in tumor cells. Oncogene 2004; 23:7611-20. [PMID: 15361826 DOI: 10.1038/sj.onc.1207970] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We previously reported that the TRAIL (tumor necrosis factor (TNF)-related apoptosis-inducing ligand)-induced death signal requires amplification by mitochondria in certain cell types, for example, in type II cells. Here, we provide for the first time evidence that the natural compound betulinic acid (BetA) cooperated with TRAIL to induce apoptosis in tumor cells. Through functional complementation, simultaneous stimulation of the death receptor pathway by TRAIL and the mitochondrial pathway by BetA resulted in complete activation of effector caspases, apoptosis and inhibition of clonogenic survival. BetA and TRAIL cooperated to trigger loss of mitochondrial membrane potential and release of cytochrome c and Smac from mitochondria. Also, combination treatment with BetA and TRAIL resulted in increased cleavage of caspase-8 and Bid indicating that activation of effector caspases may feed back in a positive amplification loop. Importantly, the combination treatment with BetA and TRAIL cooperated to induce apoptosis in different tumor cell lines and also in primary tumor cells, but not in normal human fibroblasts indicating some tumor specificity. Since most human cancers represent type II cells, triggering the mitochondrial pathway by BetA may be a novel approach to enhance the efficacy of TRAIL-based therapies, which warrants further investigation.
Collapse
Affiliation(s)
- Simone Fulda
- University Children's Hospital, Prittwitzstr. 43, D-89075 Ulm, Germany.
| | | | | |
Collapse
|
28
|
Abstract
Apoptosis, the cell's intrinsic death program, is a key regulator of tissue homeostasis. An imbalance between cell death and proliferation may result in tumor formation. Also, killing of cancer cells by cytotoxic therapies such as chemotherapy, gamma-irradiation or ligation of death receptors is predominantly mediated by triggering apoptosis in target cells. In addition to the intrinsic mitochondrial pathway, elements of death receptor signaling pathways have been implied to contribute to the efficacy of cancer therapy. Failure to undergo apoptosis in response to anticancer therapy may lead to resistance. Also, deregulated expression of death receptor pathway molecules may contribute to tumorigenesis and tumor escape from endogenous growth control. Understanding the molecular events that regulate apoptosis induced by anticancer therapy and how cancer cells evade apoptosis may provide new opportunities for pathway-based rational therapy and for drug development.
Collapse
|