1
|
Luo Y, Sheikh TMM, Li X, Yuan Y, Yao F, Wang M, Guo X, Wu J, Shafiq M, Xie Q, Jiao X. Exploring the dynamics of gut microbiota, antibiotic resistance, and chemotherapy impact in acute leukemia patients: A comprehensive metagenomic analysis. Virulence 2024; 15:2428843. [PMID: 39620486 PMCID: PMC11622590 DOI: 10.1080/21505594.2024.2428843] [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: 05/07/2024] [Revised: 08/24/2024] [Accepted: 11/06/2024] [Indexed: 12/08/2024] Open
Abstract
Leukemia poses significant challenges to its treatment, and understanding its complex pathogenesis is crucial. This study used metagenomic sequencing to investigate the interplay between chemotherapy, gut microbiota, and antibiotic resistance in patients with acute leukemia (AL). Pre- and post-chemotherapy stool samples from patients revealed alterations in microbial richness, taxa, and antibiotic resistance genes (ARGs). The analysis revealed a decreased alpha diversity, increased dispersion in post-chemotherapy samples, and changes in the abundance of specific bacteria. Key bacteria such as Enterococcus, Klebsiella, and Escherichia coli have been identified as prevalent ARG carriers. Correlation analysis between gut microbiota and blood indicators revealed potential links between microbial species and inflammatory biomarkers, including C-reactive protein (CRP) and adenosine deaminase (ADA). This study investigated the impact of antibiotic dosage on microbiota and ARGs, revealing networks connecting co-occurring ARGs with microbial species (179 nodes, 206 edges), and networks associated with ARGs and antibiotic dosages (50 nodes, 50 edges). Antibiotics such as cephamycin and sulfonamide led to multidrug-resistant Klebsiella colonization. Our analyses revealed distinct microbial profiles with Salmonella enterica elevated post-chemotherapy in NF patients and Akkermansia muciniphila elevated pre-chemotherapy. These microbial signatures could inform strategies to modulate the gut microbiome, potentially mitigating the risk of neutropenic fever in patients undergoing chemotherapy. Finally, a comprehensive analysis of KEGG modules shed light on disrupted metabolic pathways after chemotherapy, providing insights into potential targets for managing side effects. Overall, this study revealed intricate relationships between gut microbiota, chemotherapy, and antibiotic resistance, providing new insights into improving therapy and enhancing patient outcomes.
Collapse
Affiliation(s)
- Ying Luo
- Department of Laboratory Medicine, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | | | - Xin Li
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - YuMeng Yuan
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - Fen Yao
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - Meimei Wang
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - Xiaoling Guo
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - Jilong Wu
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - Muhammad Shafiq
- Research Institute of Clinical Pharmacy, Shantou University Medical College, Shantou, China
| | - Qingdong Xie
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - Xiaoyang Jiao
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| |
Collapse
|
2
|
Li J, Wang Y, Dong C, Luo L. Advancements in leukemia management: Bridging diagnosis, prognosis and nanotechnology (Review). Int J Oncol 2024; 65:112. [PMID: 39364739 PMCID: PMC11542963 DOI: 10.3892/ijo.2024.5700] [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: 04/19/2024] [Accepted: 09/19/2024] [Indexed: 10/05/2024] Open
Abstract
Leukemia is a cancer that starts in blood stem cells in the bone marrow. Today, the proper diagnosis and prognosis of leukemia are essential in mitigating the morbidity and mortality associated with this malignancy. The advent of novel biomarkers, particularly those related to minimal residual disease, has paved the way for personalized therapeutic strategies and enables the quantitative assessment of patient responses to treatment regimens. Novel diagnostic and targeted drug delivery may be helpful for the improved management of leukemia. Genetic clinical parameters, such as chromosomal abnormalities, are crucial in diagnosing and guiding treatment decisions. These genetic markers also provide valuable prognostic information, helping to predict patient outcomes and tailor personalized treatment plans. In the present review, the studies on the diagnostic and prognostic parameters of leukemia were analyzed. The prognosis of leukemia was investigated in most of the studies, and the remaining were performed on diagnosis. The clinical and laboratory prognostic parameters were the most common, followed by diagnostic hematological parameters, diagnostic blood parameter studies, and diagnostic immunological parameters. Clinical and laboratory prognostic and hematologic parameters were the most extensively studied. The methods used to diagnose and prognose the leukemia cases in these studies were predominantly clinical hematology. Numerous surface proteins and receptors, including CD45, CD27, CD29, CD38, CD27, CD123, CD56 and CD25, react similarly in various kinds of leukemia, which are ideal for targeted drug delivery. Drug delivery to leukemia cells encounters several significant obstacles, including heterogeneity, that hinder the effectiveness of treatment. Nanocarriers play a critical role in targeted drug delivery for leukemia by enhancing the precision of treatments directed at surface proteins and receptors. Additionally, they can be functionalized with targeting drugs and antibodies to target specific tissues and cells.
Collapse
Affiliation(s)
- Jingbo Li
- Department of Ultrasound Medicine, The Second Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Yingxue Wang
- Department of Ultrasound Medicine, The Second Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Chunli Dong
- Department of Critical Care Medicine, Jilin People's Hospital, Changchun, Jilin 130021, P.R. China
| | - Lifu Luo
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| |
Collapse
|
3
|
Huang X, Wu X, Wang S, Ren Y, Xu Y, Mei C, Jin J, Tong H, Qian J. Thiotepa-busulfan-fludarabine conditioning as a promising approach prior to haploidentical allogeneic hematopoietic stem cell transplantation in hematological malignancies with extramedullary involvement: Insights from a single center. Leuk Res 2024; 147:107617. [PMID: 39531954 DOI: 10.1016/j.leukres.2024.107617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/28/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Affiliation(s)
- Xianbo Huang
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China; Zhejiang Provincial Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou, China
| | - Xianhui Wu
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China; Zhejiang Provincial Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou, China
| | - Shasha Wang
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China; Zhejiang Provincial Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou, China
| | - Yanling Ren
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China; Zhejiang Provincial Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou, China
| | - Yu Xu
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China; Zhejiang Provincial Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou, China
| | - Chen Mei
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China; Zhejiang Provincial Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou, China
| | - Jie Jin
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China; Zhejiang Provincial Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou, China.
| | - Hongyan Tong
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China; Zhejiang Provincial Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou, China.
| | - Jiejing Qian
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China; Zhejiang Provincial Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou, China.
| |
Collapse
|
4
|
Jabbour EJ, Kantarjian HM, Goekbuget N, Shah BD, Chiaretti S, Park JH, Rijneveld AW, Gore L, Fleming S, Logan AC, Ribera JM, Menne TF, Mezzi K, Zaman F, Velasco K, Boissel N. Frontline Ph-negative B-cell precursor acute lymphoblastic leukemia treatment and the emerging role of blinatumomab. Blood Cancer J 2024; 14:203. [PMID: 39562780 PMCID: PMC11577064 DOI: 10.1038/s41408-024-01179-4] [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: 09/19/2024] [Revised: 10/18/2024] [Accepted: 10/22/2024] [Indexed: 11/21/2024] Open
Abstract
This narrative review seeks to summarize chemotherapeutic regimens commonly used for patients with newly diagnosed Philadelphia (Ph) chromosome-negative B-cell precursor acute lymphoblastic leukemia (BCP-ALL) in the frontline setting and to describe the latest clinical research using the bispecific T-cell-engaging immunotherapy blinatumomab in the first-line treatment setting. Current standard-of-care chemotherapeutic backbones for newly diagnosed Ph-negative BCP-ALL are based on the same overarching treatment principle: to reduce disease burden to undetectable levels and maintain lasting remission. The adult treatment landscape has progressively evolved following the adoption of pediatric-inspired regimens. However, these intense regimens are not tolerated by all, and high-risk patients still have inferior outcomes. Therefore, designing more effective and less toxic strategies remains key to further improving efficacy and safety outcomes. Overall, the treatment landscape is evolving in the frontline, and integration of blinatumomab into different standard frontline regimens may improve overall outcomes with a favorable safety profile.
Collapse
Affiliation(s)
- Elias J Jabbour
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Hagop M Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nicola Goekbuget
- Department of Medicine II, Goethe University, University Hospital, Frankfurt, Germany
| | - Bijal D Shah
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL, USA
| | - Sabina Chiaretti
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Jae H Park
- Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Lia Gore
- Children's Hospital Colorado and the University of Colorado School of Medicine, University of Colorado Cancer Center, Aurora, CO, USA
| | - Shaun Fleming
- Department of Clinical Haematology, Alfred Hospital, Melbourne, VIC, Australia, and Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, Australia
| | - Aaron C Logan
- Hematology, Blood and Marrow Transplant, and Cellular Therapy Program, Division of Hematology/Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Josep M Ribera
- ICO-Hospital Germans Trias I Pujol. Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Tobias F Menne
- The Newcastle upon Tyne Hospitals and Newcastle University, Newcastle upon Tyne, UK
| | | | | | | | - Nicolas Boissel
- Division of Hematology, EA3518 Saint-Louis Institute for Research, Saint-Louis Hospital, Paris, France
| |
Collapse
|
5
|
Khawaji ZY, Khawaji NY, Alahmadi MA, Elmoneim AA. Prediction of Response to FDA-Approved Targeted Therapy and Immunotherapy in Acute Lymphoblastic Leukemia (ALL). Curr Treat Options Oncol 2024; 25:1163-1183. [PMID: 39102166 DOI: 10.1007/s11864-024-01237-w] [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] [Accepted: 06/18/2024] [Indexed: 08/06/2024]
Abstract
OPINION STATEMENT Acute lymphoblastic leukemia (ALL) represents the predominant cancer in pediatric populations, though its occurrence in adults is relatively rare. Pre-treatment risk stratification is crucial for predicting prognosis. Important factors for assessment include patient age, white blood cell (WBC) count at diagnosis, extramedullary involvement, immunophenotype, and cytogenetic aberrations. Minimal residual disease (MRD), primarily assessed by flow cytometry following remission, plays a substantial role in guiding management plans. Over the past decade, significant advancements in ALL outcomes have been witnessed. Conventional chemotherapy has remarkably reduced mortality rates; however, its intensive nature raises safety concerns and has led to the emergence of treatment-resistant cases with recurrence of relapses. Consequently, The U.S. Food and Drug Administration (FDA) has approved several novel treatments for relapsed/refractory ALL due to their demonstrated efficacy, as indicated by improved complete remission and survival rates. These treatments include tyrosine kinase inhibitors (TKIs), the anti-CD19 monoclonal antibody blinatumomab, anti-CD22 inotuzumab ozogamicin, anti-CD20 rituximab, and chimeric antigen receptor (CAR) T-cell therapy. Identifying the variables that influence treatment decisions is a pressing necessity for tailoring therapy based on heterogeneous patient characteristics. Key predictive factors identified in various observational studies and clinical trials include prelymphodepletion disease burden, complex genetic abnormalities, and MRD. Furthermore, the development of serious adverse events following treatment could be anticipated through predictive models, allowing for appropriate prophylactic measures to be considered. The ultimate aim is to incorporate the concept of precision medicine in the field of ALL through valid prediction platform to facilitate the selection of the most suitable treatment approach.
Collapse
Affiliation(s)
| | | | | | - Abeer Abd Elmoneim
- Women and Child Health Department, Taibah University, Madinah, Kingdom of Saudi Arabia
- 2nd Affiliation: Pediatric Department, Faculty of Medicine, Sohag University, Sohag, Egypt
| |
Collapse
|
6
|
Ng HJ, Alata MK, Nguyen QT, Huynh Duc Vinh P, Tan JY, Wong CL. Managing and treating COVID-19 in patients with hematological malignancies: a narrative review and expert insights. Clin Exp Med 2024; 24:119. [PMID: 38833206 PMCID: PMC11150206 DOI: 10.1007/s10238-024-01381-5] [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: 04/08/2024] [Accepted: 05/18/2024] [Indexed: 06/06/2024]
Abstract
Patients with hematologic malignancies (HMs) are at a significantly higher risk of contracting COVID-19 and experiencing severe outcomes compared to individuals without HMs. This heightened risk is influenced by various factors, including the underlying malignancy, immunosuppressive treatments, and patient-related factors. Notably, immunosuppressive regimens commonly used for HM treatment can lead to the depletion of B cells and T cells, which is associated with increased COVID-19-related complications and mortality in these patients. As the pandemic transitions into an endemic state, it remains crucial to acknowledge and address the ongoing risk for individuals with HMs. In this review, we aim to summarize the current evidence to enhance our understanding of the impact of HMs on COVID-19 risks and outcomes, identify particularly vulnerable individuals, and emphasize the need for specialized clinical attention and management. Furthermore, the impaired immune response to COVID-19 vaccination observed in these patients underscores the importance of implementing additional mitigation strategies. This may include targeted prophylaxis and treatment with antivirals and monoclonal antibodies as indicated. To provide practical guidance and considerations, we present two illustrative cases to highlight the real-life challenges faced by physicians caring for patients with HMs, emphasizing the need for individualized management based on disease severity, type, and the unique circumstances of each patient.
Collapse
Affiliation(s)
- Heng Joo Ng
- Department of Haematology, Singapore General Hospital, Singapore, Singapore
| | | | - Quang The Nguyen
- Stem Cell Transplantation Department, Blood Transfusion Hematology Hospital, Ho Chi Minh, Vietnam
| | - Phu Huynh Duc Vinh
- Stem Cell Transplantation Department, Blood Transfusion Hematology Hospital, Ho Chi Minh, Vietnam
| | - Jing Yuan Tan
- Department of Haematology, Singapore General Hospital, Singapore, Singapore
| | - Chieh Lee Wong
- Department of Haematology, Sunway Medical Centre, Bandar Sunway, Selangor, Malaysia.
- School of Medical and Life Sciences, Sunway University, Bandar Sunway, Selangor, Malaysia.
| |
Collapse
|
7
|
Li X, Wu T, Chen W, Zhang J, Jiang Y, Deng J, Long W, Qin X, Zhou Y. Andrographolide acts with dexamethasone to inhibit the growth of acute lymphoblastic leukemia CEM‑C1 cells via the regulation of the autophagy‑dependent PI3K/AKT/mTOR signaling pathway. Biomed Rep 2024; 20:43. [PMID: 38357243 PMCID: PMC10865295 DOI: 10.3892/br.2024.1731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 08/29/2023] [Indexed: 02/16/2024] Open
Abstract
Acute lymphoblastic leukemia (ALL) is one of the most common malignant tumor types of the circulatory system. Dexamethasone (DEX) acts on the glucocorticoid (GC) receptor (GR) and is a first-line chemotherapy drug for ALL. However, long-term or high-dose applications of the drug can not only cause adverse reactions, such as osteoporosis and high blood pressure, but can also cause downregulation of GR and lead to drug resistance. In the present study, reverse transcription-quantitative PCR, western blotting and LysoTracker Red staining were used to observe the effects of DEX and andrographolide (AND; a botanical with antitumorigenic properties) combined treatment. It was found that AND enhanced the sensitivity of CEM-C1 cells, a GC-resistant cell line, to DEX, and synergistically upregulated GR both at the transcriptional and post-transcriptional level with DEX. The combination of AND with DEX synergistically alkalized lysosomal lumen and downregulated the expression of autophagy-related genes Beclin1 and microtubule-associated 1 protein light chain 3 (LC3), thereby inhibiting autophagy. Knocking down LC3 expression enhanced GR expression, suggesting that GR was regulated by autophagy. Furthermore, compared with the monotherapy group (AND or DEX in isolation), AND interacted with DEX to activate the autophagy-dependent PI3K/AKT/mTOR signaling pathway by enhancing the phosphorylation of PI3K, AKT and mTOR, thereby decreasing GR degradation and increasing the sensitivity of cells to GCs. In conclusion, the present study demonstrated that AND exhibited a synergistic anti-ALL effect with DEX via upregulation of GR, which was orchestrated by the autophagy-related PI3K/AKT/mTOR signaling pathway. The results of the present study therefore provided novel research avenues and strategies for the treatment of ALL.
Collapse
Affiliation(s)
- Xiaowen Li
- Department of Clinical Pharmacy, College of Pharmacy, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region 541199, P.R. China
| | - Tong Wu
- Department of Clinical Pharmacy, College of Pharmacy, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region 541199, P.R. China
| | - Weihong Chen
- Department of Clinical Pharmacy, College of Pharmacy, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region 541199, P.R. China
| | - Jiannan Zhang
- Department of Clinical Pharmacy, College of Pharmacy, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region 541199, P.R. China
| | - Yanping Jiang
- Department of Clinical Medicine, College of Lingui Clinical Medicine, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region 541199, P.R. China
| | - Jianzhi Deng
- Guangxi Key Laboratory of Embedded Technology and Intelligent System, Guilin University of Technology, Guilin, Guangxi Zhuang Autonomous Region 541004, P.R. China
| | - Wenqing Long
- Department of Clinical Medicine, College of Lingui Clinical Medicine, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region 541199, P.R. China
| | - Xi Qin
- Department of Medical Oncology, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region 541001, P.R. China
| | - Yuehan Zhou
- Department of Clinical Pharmacy, College of Pharmacy, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region 541199, P.R. China
| |
Collapse
|
8
|
Ramezani F, Moghadasi M, Shamsasenjan K, Narmani A. Folic Acid-Decorated Chitosan-PLGA Nanobiopolymers for Targeted Drug Delivery to Acute Lymphoblastic Leukemia Cells: In Vitro Studies. Technol Cancer Res Treat 2024; 23:15330338241308077. [PMID: 39711084 DOI: 10.1177/15330338241308077] [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] [Indexed: 12/24/2024] Open
Abstract
OBJECTIVES This study developed a drug delivery system (DDS) using folic acid (FA)-functionalized chitosan (CS) and poly (lactic-co-glycolic acid) (PLGA) nanocarriers for targeted sodium butyrate (NB) delivery to leukemia cells (NALM6). The goal was to enhance NB's therapeutic efficacy while reducing its cytotoxicity to non-malignant cells. METHODS FA-CS-PLGA nanocarriers were synthesized and characterized using Fourier-transform infrared spectroscopy (FT-IR), dynamic light scattering (DLS), zeta potential analysis, transmission electron microscopy (TEM), and thermogravimetric analysis (TGA). Encapsulation efficiency, release kinetics, cytotoxicity, and apoptosis induction were assessed using MTT assays and flow cytometry in NALM6 cells. RESULTS The FA-CS-PLGA nanocarriers had a surface charge of 34.2 ± 0.12 mV and a size range of 40-60 nm. Encapsulation efficiency was 16%, with 16% of NB released within the first 4 h. MTT assays showed a reduction in leukemia cell viability to 26% after 24 h with 400 nM FA-CS-PLGA-NB, compared to over 50% viability with pure NB. The IC50 was around 300 nM. Flow cytometry revealed that FA-CS-PLGA-NB induced apoptosis in over 20% of leukemia cells, far exceeding the 5% induced by unmodified NB. CONCLUSION FA-CS-PLGA nanocarriers show significant promise as a targeted DDS for leukemia therapy, enhancing NB delivery to leukemia cells and improving therapeutic efficacy while minimizing off-target toxicity. These results support further in vivo studies and potential clinical applications.
Collapse
Affiliation(s)
- Fatemeh Ramezani
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Moghadasi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Karim Shamsasenjan
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Asghar Narmani
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| |
Collapse
|