1
|
Al-Hamaly MA, Winter E, Blackburn JS. The mitochondria as an emerging target of self-renewal in T-cell acute lymphoblastic leukemia. Cancer Biol Ther 2025; 26:2460252. [PMID: 39905687 PMCID: PMC11801350 DOI: 10.1080/15384047.2025.2460252] [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: 05/21/2024] [Revised: 12/22/2024] [Accepted: 01/24/2025] [Indexed: 02/06/2025] Open
Abstract
Acute lymphocytic leukemia (ALL) is the most common leukemia in children, with the T-cell subtype (T-ALL) accounting for 15% of those cases. Despite advancements in the treatment of T-ALL, patients still face a dismal prognosis following their first relapse. Relapse can be attributed to the inability of chemotherapy agents to eradicate leukemia stem cells (LSC), which possess self-renewal capabilities and are responsible for the long-term maintenance of the disease. Mitochondria have been recognized as a therapeutic vulnerability for cancer stem cells, including LSCs. Mitocans have shown promise in T-ALL both in vitro and in vivo, with some currently in early-phase clinical trials. However, due to challenges in studying LSCs in T-ALL, our understanding of how mitochondrial function influences self-renewal remains limited. This review highlights the emerging literature on targeting mitochondria in diverse T-ALL models, emphasizing specific mitochondrial vulnerabilities linked to LSC self-renewal and their potential to significantly improve T-ALL treatment.
Collapse
Affiliation(s)
- Majd A. Al-Hamaly
- Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Evelyn Winter
- Department of Agriculture, Biodiversity and Forestry, Federal University of Santa Catarina, Curitibanos, Brazil
| | - Jessica S. Blackburn
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
- Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
2
|
Siew ZY, Ong GK, Wong ST, Leong PP, Tan BS, Leong CO, Chupri JB, Fang CM, Voon K. Safety profile of sikamat virus and its oncolytic potential in leukemic cells and cancer stem cells. Sci Rep 2025; 15:13817. [PMID: 40258869 PMCID: PMC12012088 DOI: 10.1038/s41598-025-96061-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/25/2025] [Indexed: 04/23/2025] Open
Abstract
Leukaemia remains a global health concern. The oncotherapy resistance of leukaemia might be due to the existence of cancer stem cell populations. This study investigated the therapeutic potential of Sikamat virus (PRV7S), a Pteropine orthoreovirus, as an oncolytic virus against acute myeloid leukaemia (AML) and chronic myeloid leukaemia (CML). Using AML and CML cell lines (THP-1 and K562), as well as an AML-M5-derived cancer stem cell (CSC) model, PRV7S was shown to infect these leukaemic cells, replicate within them, and reduce their viability. PRV7S-induced cell death was associated with caspase-mediated apoptosis without significant cell cycle arrest. Transcriptomic and proteomic analyses revealed that PRV7S infection altered several cell death pathways, including apoptosis and necroptosis, highlighting its complex cell death mechanisms. PRV7S replicated efficiently in infected cells, though it did not cause persistent infection. An in vivo safety evaluation in immunocompetent mice demonstrated that PRV7S was well-tolerated, showing no adverse effects on survival, body weight, or histopathology, and no evidence of viral persistence. These findings suggest PRV7S as a promising oncolytic candidate for myeloid leukaemia, with potential efficacy against CSCs and a favourable safety profile. In conclusion, the study provides new insights into the cellular pathways involved in PRV7S-mediated oncolysis and supports further exploration of PRV7S's potential against resistant leukaemic and solid tumours.
Collapse
MESH Headings
- Animals
- Neoplastic Stem Cells/virology
- Neoplastic Stem Cells/pathology
- Neoplastic Stem Cells/metabolism
- Humans
- Oncolytic Virotherapy/methods
- Oncolytic Viruses/physiology
- Mice
- Cell Line, Tumor
- Apoptosis
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Cell Survival
Collapse
Affiliation(s)
- Zhen Yun Siew
- School of Pharmacy, University of Nottingham Malaysia, 43500, Semenyih, Selangor, Malaysia.
| | - Ghee Khang Ong
- School of Medicine, IMU University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Siew Tung Wong
- School of Medicine, IMU University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia.
| | - Pooi Pooi Leong
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, 43000, Kajang, Selangor, Malaysia
| | - Boon Shing Tan
- AGTC Genomics, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Chee-Onn Leong
- AGTC Genomics, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Juita Binti Chupri
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
| | - Chee-Mun Fang
- School of Pharmacy, University of Nottingham Malaysia, 43500, Semenyih, Selangor, Malaysia
| | - Kenny Voon
- School of Pharmacy, University of Nottingham Malaysia, 43500, Semenyih, Selangor, Malaysia.
- School of Medicine, IMU University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia.
| |
Collapse
|
3
|
Gonçalves AC, Alves R, Sarmento-Ribeiro AB. Advancements in Biomarkers and Molecular Targets in Hematological Neoplasias. Int J Mol Sci 2024; 25:6570. [PMID: 38928276 PMCID: PMC11203996 DOI: 10.3390/ijms25126570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 06/05/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024] Open
Abstract
Hematological neoplasias are among the most common cancers worldwide, and the number of new cases has been on the rise since 1990, reaching 1 [...].
Collapse
Affiliation(s)
- Ana Cristina Gonçalves
- Laboratory of Oncobiology and Hematology (LOH), University Clinics of Hematology and Oncology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (R.A.); (A.B.S.-R.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR)—Group of Environmental Genetics of Oncobiology (CIMAGO), FMUC, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
| | - Raquel Alves
- Laboratory of Oncobiology and Hematology (LOH), University Clinics of Hematology and Oncology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (R.A.); (A.B.S.-R.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR)—Group of Environmental Genetics of Oncobiology (CIMAGO), FMUC, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
| | - Ana Bela Sarmento-Ribeiro
- Laboratory of Oncobiology and Hematology (LOH), University Clinics of Hematology and Oncology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (R.A.); (A.B.S.-R.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR)—Group of Environmental Genetics of Oncobiology (CIMAGO), FMUC, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
- Hematology Service, Centro Hospitalar Universitário de Coimbra, Unidade Local de Saúde de Coimbra, 3000-061 Coimbra, Portugal
| |
Collapse
|
4
|
Yan G, Xiao Q, Zhao J, Chen H, Xu Y, Tan M, Peng L. Brucea javanica derived exosome-like nanovesicles deliver miRNAs for cancer therapy. J Control Release 2024; 367:425-440. [PMID: 38295998 DOI: 10.1016/j.jconrel.2024.01.060] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 01/25/2024] [Accepted: 01/27/2024] [Indexed: 02/05/2024]
Abstract
Triple-negative breast cancer (TNBC) is characterized by complex heterogeneity, high recurrence and metastasis rates, and short overall survival, owing to the lack of endocrine and targeted receptors, which necessitates chemotherapy as the major treatment regimen. Exosome-like nanovesicles derived from medicinal plants have shown great potential as novel biotherapeutics for cancer therapy by delivering their incorporated nucleic acids, especially microRNAs (miRNAs), to mammalian cells. In this study, we isolated exosome-like nanovesicles derived from B. javanica (BF-Exos) and investigated their influence and underlying molecular mechanisms in TNBC. We found that BF-Exos delivered 10 functional miRNAs to 4T1 cells, significantly retarding the growth and metastasis of 4T1 cells by regulating the PI3K/Akt/mTOR signaling pathway and promoting ROS/caspase-mediated apoptosis. Moreover, BF-Exos were shown to inhibit the secretion of vascular endothelial growth factor, contributing to anti-angiogenesis in the tumor microenvironment. In vivo, BF-Exos inhibited tumor growth, metastasis, and angiogenesis in breast tumor mouse models, while maintaining high biosafety. Overall, BF-Exos are considered promising nanoplatforms for the delivery of medicinal plant-derived nucleic acids, with great potential to be developed into novel biotherapeutics for the treatment of TNBC.
Collapse
Affiliation(s)
- Ge Yan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Qiyao Xiao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Jingyu Zhao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Haoran Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Yang Xu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Minhong Tan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Lihua Peng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, PR China; Jinhua Institute of Zhejiang University, Jinhua 321299, Zhejiang, PR China.
| |
Collapse
|
5
|
Adekiya TA, Moore M, Thomas M, Lake G, Hudson T, Adesina SK. Preparation, Optimization, and In-Vitro Evaluation of Brusatol- and Docetaxel-Loaded Nanoparticles for the Treatment of Prostate Cancer. Pharmaceutics 2024; 16:114. [PMID: 38258124 PMCID: PMC10819281 DOI: 10.3390/pharmaceutics16010114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 11/30/2023] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Challenges to docetaxel use in prostate cancer treatment include several resistance mechanisms as well as toxicity. To overcome these challenges and to improve the therapeutic efficacy in heterogeneous prostate cancer, the use of multiple agents that can destroy different subpopulations of the tumor is required. Brusatol, a multitarget inhibitor, has been shown to exhibit potent anticancer activity and play an important role in drug response and chemoresistance. Thus, the combination of brusatol and docetaxel in a nanoparticle platform for the treatment of prostate cancer is expected to produce synergistic effects. In this study, we reported the development of polymeric nanoparticles for the delivery of brusatol and docetaxel in the treatment of prostate cancer. The one-factor-at-a-time method was used to screen for formulation and process variables that impacted particle size. Subsequently, factors that had modifiable effects on particle size were evaluated using a 24 full factorial statistical experimental design followed by the optimization of drug loading. The optimization of blank nanoparticles gave a formulation with a mean size of 169.1 nm ± 4.8 nm, in agreement with the predicted size of 168.333 nm. Transmission electron microscopy showed smooth spherical nanoparticles. The drug release profile showed that the encapsulated drugs were released over 24 h. Combination index data showed a synergistic interaction between the drugs. Cell cycle analysis and the evaluation of caspase activity showed differences in PC-3 and LNCaP prostate cancer cell responses to the agents. Additionally, immunoblots showed differences in survivin expression in LNCaP cells after treatment with the different agents and formulations for 24 h and 72 h. Therefore, the nanoparticles are potentially suitable for the treatment of advanced prostate cancer.
Collapse
Affiliation(s)
- Tayo Alex Adekiya
- Department of Pharmaceutical Sciences, College of Pharmacy, Howard University, Washington, DC 20059, USA
| | - Madison Moore
- Department of Biology, Howard University, Washington, DC 20059, USA
| | - Michael Thomas
- Department of Biology, Howard University, Washington, DC 20059, USA
| | - Gabriel Lake
- Department of Pharmaceutical Sciences, College of Pharmacy, Howard University, Washington, DC 20059, USA
| | - Tamaro Hudson
- Cancer Center, Howard University, Washington, DC 20059, USA
| | - Simeon K. Adesina
- Department of Pharmaceutical Sciences, College of Pharmacy, Howard University, Washington, DC 20059, USA
| |
Collapse
|
6
|
Villa-Morales M, Pérez-Gómez L, Pérez-Gómez E, López-Nieva P, Fernández-Navarro P, Santos J. Identification of NRF2 Activation as a Prognostic Biomarker in T-Cell Acute Lymphoblastic Leukaemia. Int J Mol Sci 2023; 24:10350. [PMID: 37373496 DOI: 10.3390/ijms241210350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/15/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
The standard-of-care treatment of T-cell acute lymphoblastic leukaemia (T-ALL) with chemotherapy usually achieves reasonable rates of initial complete response. However, patients who relapse or do not respond to conventional therapy show dismal outcomes, with cure rates below 10% and limited therapeutic options. To ameliorate the clinical management of these patients, it is urgent to identify biomarkers able to predict their outcomes. In this work, we investigate whether NRF2 activation constitutes a biomarker with prognostic value in T-ALL. Using transcriptomic, genomic, and clinical data, we found that T-ALL patients with high NFE2L2 levels had shorter overall survival. Our results demonstrate that the PI3K-AKT-mTOR pathway is involved in the oncogenic signalling induced by NRF2 in T-ALL. Furthermore, T-ALL patients with high NFE2L2 levels displayed genetic programs of drug resistance that may be provided by NRF2-induced biosynthesis of glutathione. Altogether, our results indicate that high levels of NFE2L2 may be a predictive biomarker of poor treatment response in T-ALL patients, which would explain the poor prognosis associated with these patients. This enhanced understanding of NRF2 biology in T-ALL may allow a more refined stratification of patients and the proposal of targeted therapies, with the ultimate goal of improving the outcome of relapsed/refractory T-ALL patients.
Collapse
Affiliation(s)
- María Villa-Morales
- Department of Biology, Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Department of Genome Dynamics and Function, Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain
- Area of Genetics and Genomics, IIS Fundación Jiménez Díaz, 28040 Madrid, Spain
- Institute for Molecular Biology-IUBM, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Laura Pérez-Gómez
- Department of Biology, Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Department of Genome Dynamics and Function, Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain
| | - Eduardo Pérez-Gómez
- Departamento de Bioquímica y Biología Molecular, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, 28041 Madrid, Spain
| | - Pilar López-Nieva
- Department of Biology, Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Department of Genome Dynamics and Function, Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain
- Area of Genetics and Genomics, IIS Fundación Jiménez Díaz, 28040 Madrid, Spain
- Institute for Molecular Biology-IUBM, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Pablo Fernández-Navarro
- Unit of Cancer and Environmental Epidemiology, Centro Nacional de Epidemiología, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Networking Biomedical Research Centre of Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain
| | - Javier Santos
- Department of Biology, Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Department of Genome Dynamics and Function, Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain
- Area of Genetics and Genomics, IIS Fundación Jiménez Díaz, 28040 Madrid, Spain
- Institute for Molecular Biology-IUBM, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| |
Collapse
|
7
|
Jorge J, Neves J, Alves R, Geraldes C, Gonçalves AC, Sarmento-Ribeiro AB. Parthenolide Induces ROS-Mediated Apoptosis in Lymphoid Malignancies. Int J Mol Sci 2023; 24:ijms24119167. [PMID: 37298119 DOI: 10.3390/ijms24119167] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/20/2023] [Accepted: 05/21/2023] [Indexed: 06/12/2023] Open
Abstract
Lymphoid malignancies are a group of highly heterogeneous diseases frequently associated with constitutive activation of the nuclear factor kappa B (NF-κB) signaling pathway. Parthenolide is a natural compound used to treat migraines and arthritis and found to act as a potent NF-κB signaling inhibitor. This study evaluated in vitro parthenolide efficacy in lymphoid neoplasms. We assessed parthenolide metabolic activity in NCI-H929 (MM), Farage (GCB-DLBCL), Raji (BL), 697 and KOPN-8 (B-ALL), and CEM and MOLT-4 (T-ALL), by resazurin assay. Cell death, cell cycle, mitochondrial membrane potential (ΔΨmit), reactive oxygen species (ROS) and reduced glutathione (GSH) levels, activated caspase-3, FAS-ligand, and phosphorylated NF-κB p65 were evaluated using flow cytometry. CMYC, TP53, GPX1, and TXRND1 expression levels were assessed using qPCR. Our results showed that parthenolide promoted a metabolic activity decrease in all cell lines in a time-, dose-, and cell-line-dependent manner. The mechanism induced by parthenolide was demonstrated to be cell line dependent. Nonetheless, parthenolide promoted cell death by apoptosis with significant ROS increase (peroxides and superoxide anion) and GSH decrease combined with a ΔΨmit reduction across all studied cell lines. Despite the need to further understand parthenolide mechanisms, parthenolide should be considered as a possible new therapeutic approach for B- and T-lymphoid malignancies.
Collapse
Affiliation(s)
- Joana Jorge
- Laboratory of Oncobiology and Hematology (LOH), University Clinic of Hematology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR)-Group of Environmental Genetics of Oncobiology (CIMAGO), FMUC, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-061 Coimbra, Portugal
| | - Joana Neves
- Laboratory of Oncobiology and Hematology (LOH), University Clinic of Hematology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Raquel Alves
- Laboratory of Oncobiology and Hematology (LOH), University Clinic of Hematology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR)-Group of Environmental Genetics of Oncobiology (CIMAGO), FMUC, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-061 Coimbra, Portugal
| | - Catarina Geraldes
- Laboratory of Oncobiology and Hematology (LOH), University Clinic of Hematology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR)-Group of Environmental Genetics of Oncobiology (CIMAGO), FMUC, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-061 Coimbra, Portugal
- Hematology Service, Centro Hospitalar e Universitário de Coimbra (CHUC), 3000-061 Coimbra, Portugal
| | - Ana Cristina Gonçalves
- Laboratory of Oncobiology and Hematology (LOH), University Clinic of Hematology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR)-Group of Environmental Genetics of Oncobiology (CIMAGO), FMUC, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-061 Coimbra, Portugal
| | - Ana Bela Sarmento-Ribeiro
- Laboratory of Oncobiology and Hematology (LOH), University Clinic of Hematology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR)-Group of Environmental Genetics of Oncobiology (CIMAGO), FMUC, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-061 Coimbra, Portugal
- Hematology Service, Centro Hospitalar e Universitário de Coimbra (CHUC), 3000-061 Coimbra, Portugal
| |
Collapse
|