1
|
Muratoğlu B, Özdemir C, Eylem CC, Reçber T, Nemutlu E, Yet İ, Uçkan-Çetinkaya D. Circadian rhythm and aryl hydrocarbon receptor crosstalk in bone marrow adipose tissue and implications in leukemia. Sci Rep 2025; 15:16387. [PMID: 40350529 PMCID: PMC12066725 DOI: 10.1038/s41598-025-93169-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 03/05/2025] [Indexed: 05/14/2025] Open
Abstract
Leukemic cells modulate the bone marrow microenvironment to enhance their survival. Lipolysis in bone marrow adipose tissue (BMAT) has emerged as a critical factor supporting leukemic cell survival, yet understanding its primary role in leukemia development remains limited. Fanconi anemia (FA), characterized by a predisposition to acute myeloid leukemia (AML) and hypersensitivity to environmental toxins, is a transitional model for studying leukemic transformation. İntegrated multi-omics analyses were conducted on BMAT-derived mesenchymal stem/stromal cells (MSCs) from healthy donors (HD), AML, and FA patients. These analyses revealed intricate interactions among genes, metabolites, and lipids. Particularly noteworthy were the effects observed following the inhibition of aryl hydrocarbon receptor (AhR) signaling by StemRegenin1 (SR1). BMAT-MSCs showed increased expression of epithelial-mesenchymal transition (EMT) genes in FA and AML, suggesting a potential shift towards cancer-associated fibroblasts in the dysregulated marrow microenvironment. Identification of potential circadian rhythm biomarkers (NPAS2, PER2, BHLHE40, PER3, CIART) in BMAT-MSCs indicates a link between related lipid metabolism genes (e.g., PTGS1, PIK3R1) and SR1 treatment, implicating them in lipolysis processes. Dysregulation of circadian rhythm-related genes (CIART, BHLHE40, NPAS2) in AML BMAT-MSCs, along with changes in circulating lipid metabolites like palmitate suggests their role in shaping the leukemia microenvironment. Upregulation of FABP5 and CD36 suggests a novel molecular mechanism involving FABP5 in AhR-mediated circadian regulation and identifies CD36 as a potential partner for FABP5 in BMAT-MSCs. Overall, this study unveils the interplay between AhR signaling, circadian rhythm, and the leukemia microenvironment in BMAT-MSCs, offering new insights into leukemia pathogenesis and therapeutic opportunities.
Collapse
Affiliation(s)
- Bihter Muratoğlu
- Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, 06100, Sihhiye, Ankara, Turkey
- Department of Stem Cell Sciences, Institute of Health Sciences, Hacettepe University, 06100, Sihhiye, Ankara, Turkey
| | - Cansu Özdemir
- Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, 06100, Sihhiye, Ankara, Turkey.
- Department of Stem Cell Sciences, Institute of Health Sciences, Hacettepe University, 06100, Sihhiye, Ankara, Turkey.
| | - Cemil Can Eylem
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, 06100, Sihhiye, Ankara, Turkey
| | - Tuba Reçber
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, 06100, Sihhiye, Ankara, Turkey
| | - Emirhan Nemutlu
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, 06100, Sihhiye, Ankara, Turkey
| | - İdil Yet
- Department of Bioinformatics, Institute of Health Sciences, Hacettepe University, 06100, Sihhiye, Ankara, Turkey
| | - Duygu Uçkan-Çetinkaya
- Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, 06100, Sihhiye, Ankara, Turkey.
- Department of Stem Cell Sciences, Institute of Health Sciences, Hacettepe University, 06100, Sihhiye, Ankara, Turkey.
- Division of Hematology, Department of Pediatrics, Hacettepe University Faculty of Medicine, 06100, Sihhiye, Ankara, Turkey.
| |
Collapse
|
2
|
Hirschbühl K, Märkl B, Müller G, Schaller T, Claus R, Sommer S, Schmutz M, Trepel M, Schmid C, Dintner S. Molecular Genetic Analysis of Bone Marrow Core Biopsy as an Alternative or Adjunct to Bone Marrow Aspirate and/or Peripheral Blood in Hematologic Myeloid Neoplasms. Diagnostics (Basel) 2025; 15:991. [PMID: 40310376 PMCID: PMC12025941 DOI: 10.3390/diagnostics15080991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/04/2025] [Accepted: 04/09/2025] [Indexed: 05/02/2025] Open
Abstract
Background: The diagnosis of hematologic neoplasms is usually based on a synopsis of the peripheral blood (PB) and bone marrow findings. Morphology continues to be the cornerstone, but genetic analysis plays an increasingly important role. In routine workup, molecular genetic analysis is performed from a bone marrow aspirate (BMA). In the event of inadequate aspiration, PB is used. Not infrequently, however, PB only partially represents the disease. In this situation, molecular genetic analysis of formalin-fixed and paraffin-embedded (FFPE) bone marrow core biopsy (BMCB) could be a better alternative than PB. However, no systematic correlation of genetic findings from BMCB with results from BMA and PB has been reported. Methods: Therefore, BMCB obtained during routine diagnostics were subjected to post hoc molecular genetic analysis using next generation sequencing (NGS). The identified molecular genetic alterations were then compared with data within routine diagnostics of the corresponding BMA and/or PB. Results: In total, 29 BMCB and corresponding BMA samples were analyzed, and in 12/29 cases PB was additionally available. The analysis of BMCB and BMA showed identical results in 17 cases, but BMCB revealed a gain of information in 11, while in only 1 case, BMCB failed to identify the mutations in comparison to BMA. Conclusions: Despite the small numbers, molecular genetic analysis of bone marrow core biopsy using next generation sequencing could detect relevant additional gene mutations compared to bone marrow aspirate and/or peripheral blood.
Collapse
Affiliation(s)
- Klaus Hirschbühl
- Hematology and Oncology, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany; (R.C.); (S.S.); (M.S.); (M.T.); (C.S.)
- Bavarian Center for Cancer Research (BZKF), 86156 Augsburg, Germany; (B.M.); (T.S.); (S.D.)
| | - Bruno Märkl
- Bavarian Center for Cancer Research (BZKF), 86156 Augsburg, Germany; (B.M.); (T.S.); (S.D.)
- Pathology, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany
| | - Gernot Müller
- Department of Computational Statistics and Data Analysis, University of Augsburg, 86159 Augsburg, Germany;
| | - Tina Schaller
- Bavarian Center for Cancer Research (BZKF), 86156 Augsburg, Germany; (B.M.); (T.S.); (S.D.)
- Pathology, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany
| | - Rainer Claus
- Hematology and Oncology, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany; (R.C.); (S.S.); (M.S.); (M.T.); (C.S.)
- Bavarian Center for Cancer Research (BZKF), 86156 Augsburg, Germany; (B.M.); (T.S.); (S.D.)
- Pathology, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany
| | - Sebastian Sommer
- Hematology and Oncology, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany; (R.C.); (S.S.); (M.S.); (M.T.); (C.S.)
- Onkologie/Hämatologie im Elisenhof, 80335 München, Germany
| | - Maximilian Schmutz
- Hematology and Oncology, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany; (R.C.); (S.S.); (M.S.); (M.T.); (C.S.)
- Bavarian Center for Cancer Research (BZKF), 86156 Augsburg, Germany; (B.M.); (T.S.); (S.D.)
| | - Martin Trepel
- Hematology and Oncology, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany; (R.C.); (S.S.); (M.S.); (M.T.); (C.S.)
- Bavarian Center for Cancer Research (BZKF), 86156 Augsburg, Germany; (B.M.); (T.S.); (S.D.)
| | - Christoph Schmid
- Hematology and Oncology, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany; (R.C.); (S.S.); (M.S.); (M.T.); (C.S.)
- Bavarian Center for Cancer Research (BZKF), 86156 Augsburg, Germany; (B.M.); (T.S.); (S.D.)
| | - Sebastian Dintner
- Bavarian Center for Cancer Research (BZKF), 86156 Augsburg, Germany; (B.M.); (T.S.); (S.D.)
- Pathology, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany
| |
Collapse
|
3
|
Azevedo PL, Rezende M, Felix M, Corrêa S, Abdelhay E, Binato R. SAA1 Protein: A Potential Biomarker for Acute Myeloid Leukemia. Biomedicines 2025; 13:880. [PMID: 40299483 PMCID: PMC12024993 DOI: 10.3390/biomedicines13040880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/07/2025] [Accepted: 03/09/2025] [Indexed: 04/30/2025] Open
Abstract
Background/Objectives: Despite its heterogeneity and diagnostic challenges, acute myeloid leukemia (AML) originates from stem cell transformation and alterations in the hematopoietic niche (HN) could be related to leukemic transformation. Therefore, the aim of this study was to evaluate the protein profile of HN from AML patients and compare it with the profile of healthy donors (HDs). Methods: A proteomic analysis was conducted to identify differentially expressed (DE) proteins in BM plasma from AML patients and HD. In silico analysis was performed to identify biological processes and signaling pathways involved. Additionally, ELISA confirmed the expression of the DE protein of interest in BM plasma samples. Results: Proteomic analysis revealed alterations in the plasma profiles of AML patients and 36 DE proteins were found. Among then, we highlight C8G, CFB, SAA1, SERPINA3 and SERPINC1, which are related to inflammatory response process. Thus, considering the role of the secreted protein SAA1 in the inflammatory context and that it is described as a potential biomarker in several tumors, we selected SAA1 for ELISA confirmation. The results corroborated our findings, indicating that increased expression of SAA1 could be related to AML. Our results also revealed that SAA1 can stimulate immune signaling through NF-kappa-B activation. Conclusions: These findings position SAA1 as a promising biomarker for AML diagnosis, offering a potential tool for more accurate identification of the disease. Nevertheless, further studies are needed to understand the relationship of SAA1 with the leukemic transformation process in AML and its potential clinical use.
Collapse
|
4
|
Hajipirloo LK, Nabigol M, Khayami R, Karami N, Farsani MA, Navidinia AA. Construction of a stromal-related prognostic model in acute myeloid leukemia by comprehensive bioinformatics analysis. Curr Res Transl Med 2025; 73:103492. [PMID: 39818173 DOI: 10.1016/j.retram.2025.103492] [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: 02/09/2024] [Revised: 12/10/2024] [Accepted: 01/04/2025] [Indexed: 01/18/2025]
Abstract
BACKGROUND Stromal cells play a pivotal role in the tumor microenvironment (TME), significantly impacting the progression of acute myeloid leukemia (AML). This study sought to develop a stromal-related prognostic model for AML, aiming to uncover novel prognostic markers and therapeutic targets. METHODS RNA expression data and clinical profiles of AML patients were retrieved from the Cancer Genome Atlas (TCGA). The extent of stromal cell infiltration within the TME was quantified using the ESTIMATE algorithm. Associations between stromal scores and the French-American-British (FAB) classification, overall survival (OS), and the Cancer and Leukemia Group B (CALGB) cytogenetic risk categories were analyzed. Differentially expressed genes (DEGs) were identified, and gene ontology (GO) and protein-protein interaction (PPI) networks were constructed. Prognostic DEGs were selected through LASSO-cox regression analysis. A risk score model was then developed based on these DEGs. A stromal-related prognostic model (SPM) was constructed from the patients' risk scores (RS), and its efficacy was evaluated using Receiver Operating Characteristic (ROC) curves and a nomogram. The association between FAB, CALGB, age, and common mutations and SPM was also assessed. Ultimately, the SPM was validated using an external dataset from 246 patients in the TARGET-AML study. RESULTS Kaplan-Meier analysis revealed a significant association between stromal scores and patient survival (p = 0.04). LASSOCox regression identified four genes (MAP7D2, CDRT1, HOXB9, and IRX5) as highly predictive of survival. The prognostic model showed a strong correlation with overall survival, with higher scores indicating poorer outcomes (p = 1.48e-07). Older patients (over 60 years) faced significantly worse prognoses (p = 0.0055). Although no significant association was found between the SPM and the FAB classification (p = 0.063), both poor and intermediate/normal cytogenetic groups had significantly higher SPM risk scores than the favorable group (p = 0.0057 and 0.0026). External validation of the SPM in the TARGET-AML dataset confirmed a significant association with survival (p = 0.00035), with the area under the curve (AUC) for 10-year survival at 75.81 %. CONCLUSION Our research successfully established a stromal-related prognostic model in AML, offering new perspectives for prognostic evaluation and identifying potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Laya Khodayi Hajipirloo
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Nabigol
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Khayami
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Najibe Karami
- Hematology-Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Allahbakhshian Farsani
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Abbas Navidinia
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Hematology-Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Wu J, Li X, Liu Y, Chen G, Li R, Jiang H, Yin W, Tong X, Cao R, Wang X, Liu X, Zhou F. MMP14 from BM-MSCs facilitates progression and Ara-C resistance in acute myeloid leukemia via the JAK/STAT pathway. Exp Hematol Oncol 2025; 14:43. [PMID: 40121502 PMCID: PMC11929205 DOI: 10.1186/s40164-025-00635-6] [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: 09/16/2024] [Accepted: 03/07/2025] [Indexed: 03/25/2025] Open
Abstract
Growing evidence underscores the pivotal impact of crosstalk between leukemic stem cells (LSCs) and mesenchymal stromal cells (MSCs) within their niche on leukemia initiation, progression, and therapy response. Although MMP14 plays an important role in inflammation and cancer, the regulation and role of MSC-derived MMP14 in acute myeloid leukemia (AML) are largely unknown. Here, we found that AML patient-derived MSCs (AML-MSCs) were more supportive of AML cell growth compared to healthy donor-derived MSCs (HD-MSCs). Moreover, AML-MSCs and HD-MSCs showed significant differences in gene expression and protein expression profiles. Knockdown of MMP14 in MSCs inhibited the CFU-F ability of MSC cells and increased the proportion of cells in the G0 phase, thereby inhibiting proliferation. Co-culture with MSCs inhibited the proliferation and cell cycle progression of leukemia cells, while increasing the apoptosis rate, thus impairing the leukemogenic potential of AML cells both in vitro and in vivo. Mechanistic studies revealed that MMP14-mediated alterations in the AML stromal microenvironment are driven by PGE2 secretion and activation of the JAK-STAT pathway, promoting leukemia progression. Notably, inhibition of MMP14 can attenuate the chemotherapy resistance of AML cells induced by MSCs to cytarabine (Ara-C). Together, our study, for the first time, demonstrates the critical role of MSC-derived MMP14 in promoting AML progression and chemoresistance. Targeting MMP14 signaling pathways may offer novel therapeutic options for AML.
Collapse
Affiliation(s)
- Jinxian Wu
- Department of Hematology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430072, P. R. China
| | - Xinqi Li
- Department of Hematology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430072, P. R. China
| | - Yin Liu
- Department of Hematology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430072, P. R. China
| | - Guopeng Chen
- Department of Hematology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430072, P. R. China
| | - Ruihang Li
- Department of Hematology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430072, P. R. China
| | - Hongqiang Jiang
- Department of Hematology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430072, P. R. China
| | - Wanyue Yin
- Department of Hematology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430072, P. R. China
| | - Xiqin Tong
- Department of Hematology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430072, P. R. China
| | - Rui Cao
- Department of Hematology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430072, P. R. China
| | - Xianwang Wang
- Department of Biochemistry and Molecular Biology, Center for Molecular Medicine, Health Science Center, Yangtze University, Jingzhou, 434023, Hubei, China
- Shannan Maternal and Child Health Hospital, Shannan, Xizang, 856100, China
| | - Xiaoyan Liu
- Department of Hematology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430072, P. R. China.
| | - Fuling Zhou
- Department of Hematology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430072, P. R. China.
- Research Center for Lifespan Health, Wuhan University, Wuhan, 430072, Hubei, China.
| |
Collapse
|
6
|
Chai Z, Yuan Z, Chen Y. Stem cell status and prognostic applications of cuproptosis-associated lncRNAs in acute myeloid leukemia. Front Cell Dev Biol 2025; 12:1549294. [PMID: 39877157 PMCID: PMC11772438 DOI: 10.3389/fcell.2024.1549294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025] Open
Abstract
Introduction Acute myeloid leukemia (AML), a highly heterogeneous hematological malignancy, remains a major challenge in adult oncology. Stem cell research has highlighted the crucial role of long noncoding RNA (lncRNA) in regulating cellular differentiation and self-renewal processes, which are pivotal in AML pathogenesis and therapy resistance. Methods This study explores the relationship between cuproptosis-related lncRNAs and AML prognosis, providing novel insights into their impact on hematopoietic stem and progenitor cells. Results We collected clinical information from 214 AML patients in our center and analyzed the association between granulocyte recovery after chemotherapy, cuproptosis, and prognosis. Additionally, we developed a prognostic model-the cuproptosis-associated long noncoding RNA prognostic model (CRLPM)-y analyzing data from The Cancer Genome Atlas (TCGA). Patients were stratified into high- and low-risk groups based on CRLPM, revealing significant survival differences. High-risk patients demonstrated lower sensitivity to chemotherapeutic agents such as Axitinib, GSK429286A, Navitoclax, and ZM-447439, underscoring the need for alternative therapeutic strategies. Discussion CRLPM offers a promising framework for integrating stem cell-focused approaches into personalized treatment regimens, paving the way for precision medicine in AML management.
Collapse
Affiliation(s)
- Zhuodong Chai
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, TX, United States
| | - Zhongyue Yuan
- Department of Industrial and Molecular Pharmaceutics, Purdue University, West Lafayette, IN, United States
| | - Yifei Chen
- Department of Hematology, Jiangdu People’s Hospital, Yangzhou, China
| |
Collapse
|
7
|
Mattioda C, Voena C, Ciardelli G, Mattu C. In Vitro 3D Models of Haematological Malignancies: Current Trends and the Road Ahead? Cells 2025; 14:38. [PMID: 39791739 PMCID: PMC11720277 DOI: 10.3390/cells14010038] [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: 11/26/2024] [Revised: 12/23/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025] Open
Abstract
Haematological malignancies comprise a diverse group of life-threatening systemic diseases, including leukaemia, lymphoma, and multiple myeloma. Currently available therapies, including chemotherapy, immunotherapy, and CAR-T cells, are often associated with important side effects and with the development of drug resistance and, consequently, disease relapse. In the last decades, it was largely demonstrated that the tumor microenvironment significantly affects cancer cell proliferation and tumor response to treatment. The development of biomimetic, in vitro models may promote the investigation of the interactions between cancer cells and the tumor microenvironment and may help to better understand the mechanisms leading to drug resistance. Although advanced in vitro models have been largely explored in the field of solid tumors, due to the complex nature of the blood cancer tumor microenvironment, the mimicking of haematological malignancies mostly relies on simpler systems, often limited to two-dimensional cell culture, which intrinsically excludes the microenvironmental niche, or to ethically debated animal models. This review aims at reporting an updated overview of state-of-the-art hematological malignancies 3D in vitro models, emphasizing the key features and limitations of existing systems to inspire further research in this underexplored field.
Collapse
Affiliation(s)
- Carlotta Mattioda
- DIMEAS, Politecnico di Torino, C.so Duca degli Abruzzi 24, 10129 Torino, Italy; (C.M.); (G.C.)
| | - Claudia Voena
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Torino, Italy;
| | - Gianluca Ciardelli
- DIMEAS, Politecnico di Torino, C.so Duca degli Abruzzi 24, 10129 Torino, Italy; (C.M.); (G.C.)
| | - Clara Mattu
- DIMEAS, Politecnico di Torino, C.so Duca degli Abruzzi 24, 10129 Torino, Italy; (C.M.); (G.C.)
| |
Collapse
|
8
|
Bouzriba C, Chavez Alvarez AC, Ouellette V, Gagné-Boulet M, Hamel-Côté G, Bastien D, Laverdière I, Fortin S. N-Phenyl ureidobenzenesulfonates, a novel class of human dihydroorotate dehydrogenase inhibitors inducing differentiation and apoptosis in acute myeloid leukemia cells. Biomed Pharmacother 2024; 181:117717. [PMID: 39637752 DOI: 10.1016/j.biopha.2024.117717] [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: 08/23/2024] [Revised: 11/11/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024] Open
Abstract
N-Phenyl ureidobenzensulfonates (PUB-SOs) are a novel family of dihydroorotate dehydrogenase (DHODH) inhibitors. Herein, we investigate the potential of PUB-SOs to induce acute myeloid leukemia (AML) cell differentiation and apoptosis. To that end, we screened our chemolibrary to select the most potent PUB-SOs based on their antiproliferative activity and their ability to arrest the cell progression of AML cells in the S phase. The most promising PUB-SOs show antiproliferative activity in the range of 0.13-23 µM against THP-1, MOLM-13 and HL-60 AML cells. Moreover, those PUB-SOs arrested the cell cycle progression in the S phase. In addition, molecular docking studies evidenced their potential to bind in the brequinar-binding site located on DHODH which was confirmed using the DHODH inhibition assay showing that PUB-SOs are potent DHODH inhibitors (half maximal inhibitory concentration (IC50) = 7.7-1000 nM). Our results also show that selected PUB-SOs induced the differentiation of THP-1 and HL-60 cells into cluster of differentiation (CD) 11b+/CD14+ phenotypes, up to 74 % and 50 %, respectively. They also promoted CD11b+ differentiation in MOLM-13 cells (up to 44 %). Additionally, the prototypical PUB-SOs SFOM-0046 induced lactate dehydrogenase (LDH) release, mitochondrial stress and mitochondrial membrane potential loss in MOLM-13 cell line. Furthermore, SFOM-0046 induced apoptosis in MOLM-13 cells, which was confirmed by the increase of annexin V/propidium iodide (PI) and caspase 3/7 positive cells. In summary, our results highlight PUB-SOs as a novel family of DHODH inhibitors inducing both cell differentiation and apoptosis in AML cells, underscoring their potential as therapeutic agents for AML treatment.
Collapse
Affiliation(s)
- Chahrazed Bouzriba
- Faculté de pharmacie, Université Laval, Pavillon Ferdinand-Vandry, 1050 avenue de la Médecine, Québec, QC G1V 0A6, Canada; Centre de recherche du CHU de Québec-Université Laval, Axe oncologie, Hôpital Saint-François d'Assise, 10 rue de l'Espinay, Québec, QC G1L 3L5, Canada.
| | - Atziri Corin Chavez Alvarez
- Faculté de pharmacie, Université Laval, Pavillon Ferdinand-Vandry, 1050 avenue de la Médecine, Québec, QC G1V 0A6, Canada; Centre de recherche du CHU de Québec-Université Laval, Axe oncologie, Hôpital Saint-François d'Assise, 10 rue de l'Espinay, Québec, QC G1L 3L5, Canada; Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, 2725 Chemin Ste-Foy, Québec, QC G1V 4G5, Canada
| | - Vincent Ouellette
- Faculté de pharmacie, Université Laval, Pavillon Ferdinand-Vandry, 1050 avenue de la Médecine, Québec, QC G1V 0A6, Canada; Centre de recherche du CHU de Québec-Université Laval, Axe oncologie, Hôpital Saint-François d'Assise, 10 rue de l'Espinay, Québec, QC G1L 3L5, Canada
| | - Mathieu Gagné-Boulet
- Faculté de pharmacie, Université Laval, Pavillon Ferdinand-Vandry, 1050 avenue de la Médecine, Québec, QC G1V 0A6, Canada; Centre de recherche du CHU de Québec-Université Laval, Axe oncologie, Hôpital Saint-François d'Assise, 10 rue de l'Espinay, Québec, QC G1L 3L5, Canada
| | - Geneviève Hamel-Côté
- Centre de recherche du CHU de Québec-Université Laval, Axe oncologie, Hôpital Saint-François d'Assise, 10 rue de l'Espinay, Québec, QC G1L 3L5, Canada
| | - Dominic Bastien
- Centre de recherche du CHU de Québec-Université Laval, Axe oncologie, Centre hospitalier de l'Université Laval CHUL, 2705 Blvd Laurier, Québec, QC G1V 4G2, Canada
| | - Isabelle Laverdière
- Faculté de pharmacie, Université Laval, Pavillon Ferdinand-Vandry, 1050 avenue de la Médecine, Québec, QC G1V 0A6, Canada; Centre de recherche du CHU de Québec-Université Laval, Axe oncologie, Centre hospitalier de l'Université Laval CHUL, 2705 Blvd Laurier, Québec, QC G1V 4G2, Canada
| | - Sébastien Fortin
- Faculté de pharmacie, Université Laval, Pavillon Ferdinand-Vandry, 1050 avenue de la Médecine, Québec, QC G1V 0A6, Canada; Centre de recherche du CHU de Québec-Université Laval, Axe oncologie, Hôpital Saint-François d'Assise, 10 rue de l'Espinay, Québec, QC G1L 3L5, Canada.
| |
Collapse
|
9
|
Deng J, Tan Y, Xu Z, Wang H. Advances in hematopoietic stem cells ex vivo expansion associated with bone marrow niche. Ann Hematol 2024; 103:5035-5057. [PMID: 38684510 DOI: 10.1007/s00277-024-05773-1] [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: 10/08/2023] [Accepted: 04/19/2024] [Indexed: 05/02/2024]
Abstract
Hematopoietic stem cells (HSCs) are an ideal source for the treatment of many hematological diseases and malignancies, as well as diseases of other systems, because of their two important features, self-renewal and multipotential differentiation, which have the ability to rebuild the blood system and immune system of the body. However, so far, the insufficient number of available HSCs, whether from bone marrow (BM), mobilized peripheral blood or umbilical cord blood, is still the main restricting factor for the clinical application. Therefore, strategies to expand HSCs numbers and maintain HSCs functions through ex vivo culture are urgently required. In this review, we outline the basic biology characteristics of HSCs, and focus on the regulatory factors in BM niche affecting the functions of HSCs. Then, we introduce several representative strategies used for HSCs from these three sources ex vivo expansion associated with BM niche. These findings have deepened our understanding of the mechanisms by which HSCs balance self-renewal and differentiation and provided a theoretical basis for the efficient clinical HSCs expansion.
Collapse
Affiliation(s)
- Ju Deng
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- The Key Laboratory of Molecular Diagnosis and Treatment of Hematological Disease of Shanxi Province, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanhong Tan
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- The Key Laboratory of Molecular Diagnosis and Treatment of Hematological Disease of Shanxi Province, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Zhifang Xu
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- The Key Laboratory of Molecular Diagnosis and Treatment of Hematological Disease of Shanxi Province, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Hongwei Wang
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.
- The Key Laboratory of Molecular Diagnosis and Treatment of Hematological Disease of Shanxi Province, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
10
|
do Nascimento MC, Pereira-Martins DA, Machado-Neto JA, Rego EM. Acute myeloid leukemia-derived bone marrow mesenchymal cells exhibit improved support for leukemic cell proliferation. Hematol Transfus Cell Ther 2024; 46 Suppl 6:S48-S52. [PMID: 38307829 PMCID: PMC11726071 DOI: 10.1016/j.htct.2023.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/23/2023] [Indexed: 02/04/2024] Open
Abstract
INTRODUCTION The bone marrow (BM) microenvironment plays a significant role in acute myeloid leukemia (AML) genesis and there is evidence that BM mesenchymal stromal cells (BMMSCs) can support leukemia progenitor cell proliferation and survival and provide resistance to cytotoxic therapies. HYPOTHESIS AND METHOD Nevertheless, currently unknown are the relevance of the spatial localization of AML cells relative to the BMMSCs and whether BMMSCs from patients with AML and healthy subjects have similar properties. To address these issues, we performed a differential gene expression analysis using RNA-sequencing data generated from healthy donors (HDs) and leukemic BMMSCs. RESULTS The Gene Set Enrichment Analysis (GSEA) revealed that leukemic BMMSCs were associated with the terms "positive regulation of cell cycle", "angiogenesis" and "signaling by the estimated glomerular filtration rate (eGFR)", whereas healthy donor (HD)-derived BMMSCs were associated with "programmed cell death in response to the reactive oxygen species (ROS)", "negative regulation of the cytochrome C from the mitochondria" and "interferon signaling". Next, we evaluated the mitochondrial superoxide production in AML cells in a co-culture layered model. The superoxide production was reduced in leukemic cells in close contact (adhered to the surface or beneath the cell layer) with BMMSCs, indicating lower oxidative stress. CONCLUSION Taken together, our results suggest that AML-derived BMMSCs are transcriptionally rewired and can reduce the metabolic stress of leukemic cells.
Collapse
Affiliation(s)
- Mariane Cristina do Nascimento
- Center for Cell-Based Therapy, Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil; Hematology Division, LIM31, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - Diego A Pereira-Martins
- Center for Cell-Based Therapy, Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil; Hematology Division, LIM31, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | | | - Eduardo M Rego
- Center for Cell-Based Therapy, Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil; Hematology Division, LIM31, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil.
| |
Collapse
|
11
|
Farokhi-Fard A, Rahmati S, Hashemi Aval NS, Barkhordari F, Bayat E, Komijani S, Aghamirza Moghim Aliabadi H, Davami F. Anti-IL-1RAP scFv-mSA-S19-TAT fusion carrier as a multifunctional platform for versatile delivery of biotinylated payloads to myeloid leukemia cells. Sci Rep 2024; 14:25080. [PMID: 39443595 PMCID: PMC11500005 DOI: 10.1038/s41598-024-76851-7] [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: 08/16/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive blood cancer with frequently poor clinical outcomes. This heterogeneous malignancy encompasses genetically, molecularly, and even clinically different subgroups. This makes it difficult to develop therapeutic agents that are effective for all subtypes of the disease. Therefore, a selective, universal, and adaptable delivery platform capable of carrying various types of anti-neoplastic agents is an unmet requirement in this area. Two multifunctional fusion proteins were designed for the delivery of biotinylated cargoes to human myeloid leukemia cells by fusing an anti-IL-1RAP single-chain antibody with streptavidin (tetramer or monomer), a cell-penetrating peptide (CPP), and an endosomolytic peptide in a single biomacromolecule. The designed fusions were analyzed primarily in silico, and the biofunctionality of the selected fusion was fully characterized via several binding assays, hemolysis assay, confocal microscopy and cell cytotoxicity assay after production via the Escherichia coli (E. coli) system. The refolded protein exhibited desirable binding activity to leukemic cells, pure antigen and biotinylated BSA. Further analyses revealed efficient cellular uptake, endosomolytic activity, and nuclear penetration without any detectable cytotoxicity toward normal epithelial cells. The described platform seems to have great potential for targeted delivery of different therapeutics to malignant myeloid cells.
Collapse
MESH Headings
- Humans
- Single-Chain Antibodies/immunology
- Single-Chain Antibodies/genetics
- Recombinant Fusion Proteins/genetics
- Biotinylation
- Cell-Penetrating Peptides/chemistry
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/therapy
- Cell Line, Tumor
- Drug Delivery Systems
- Streptavidin/chemistry
- Drug Carriers/chemistry
- Leukemia, Myeloid/immunology
- Leukemia, Myeloid/drug therapy
- Leukemia, Myeloid/therapy
Collapse
Affiliation(s)
- Aref Farokhi-Fard
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Saman Rahmati
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | | | | | - Elham Bayat
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Samira Komijani
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | | | - Fatemeh Davami
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
- Pasteur Institute of Iran, No. 69, Pasteur Ave, Tehran, Iran.
| |
Collapse
|
12
|
Mesaros O, Onciul M, Matei E, Joldes C, Jimbu L, Neaga A, Serban O, Zdrenghea M, Nanut AM. Macrophages as Potential Therapeutic Targets in Acute Myeloid Leukemia. Biomedicines 2024; 12:2306. [PMID: 39457618 PMCID: PMC11505058 DOI: 10.3390/biomedicines12102306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/15/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
Acute myeloid leukemia (AML) is a heterogenous malignant hemopathy, and although new drugs have emerged recently, current treatment options still show limited efficacy. Therapy resistance remains a major concern due to its contribution to treatment failure, disease relapse, and increased mortality among patients. The underlying mechanisms of resistance to therapy are not fully understood, and it is crucial to address this challenge to improve therapy. Macrophages are immune cells found within the bone marrow microenvironment (BMME), of critical importance for leukemia development and progression. One defining feature of macrophages is their plasticity, which allows them to adapt to the variations in the microenvironment. While this adaptability is advantageous during wound healing, it can also be exploited in cancer scenarios. Thus, clinical and preclinical investigations that target macrophages as a therapeutic strategy appear promising. Existing research indicates that targeting macrophages could enhance the effectiveness of current AML treatments. This review addresses the importance of macrophages as therapeutic targets including relevant drugs investigated in clinical trials such as pexidartinib, magrolimab or bexmarilimab, but also provides new insights into lesser-known therapies, like macrophage receptor with a collagenous structure (MACRO) inhibitors and Toll-like receptor (TLR) agonists.
Collapse
Affiliation(s)
- Oana Mesaros
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Str., 400015 Cluj-Napoca, Romania
| | - Madalina Onciul
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania
| | - Emilia Matei
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania
- Department of Pathology, Ion Chiricuta Oncology Institute, 34-36 Republicii Str., 400015 Cluj-Napoca, Romania
| | - Corina Joldes
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania
- Octavian Fodor” Regional Institute of Gastroenterology and Hepatology, 19-21 Croitorilor Str., 400162 Cluj-Napoca, Romania
| | - Laura Jimbu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Str., 400015 Cluj-Napoca, Romania
| | - Alexandra Neaga
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Str., 400015 Cluj-Napoca, Romania
| | - Oana Serban
- Regina Maria” Regional Laboratory in Cluj-Napoca, 109 Observatorului Str., 400363 Cluj-Napoca, Romania
| | - Mihnea Zdrenghea
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Str., 400015 Cluj-Napoca, Romania
| | - Ana Maria Nanut
- Regina Maria” Regional Laboratory in Cluj-Napoca, 34-36 Republicii Str., 400015 Cluj-Napoca, Romania
| |
Collapse
|
13
|
Rafiq N, Khan MH, Sahibzada M, Khan SA, Syamprabha Vijayan A, Ullah N, Koodarath C, Khalil H, Ali UA, Saleem F, Almounjed S, Khaliq I. Recent Developments and Challenges in the Treatment of Acute Leukemia and Myelodysplastic Syndromes: A Systematic Review. Cureus 2024; 16:e72599. [PMID: 39610611 PMCID: PMC11604246 DOI: 10.7759/cureus.72599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2024] [Indexed: 11/30/2024] Open
Abstract
The diagnosis of acute leukemia (AL) and myelodysplastic syndrome (MDS) is critical due to their rapid progression and varied survival rates between children and older adults. These diseases are associated with significant mortality, highlighting the need for strategies to reduce the global burden of AL and MDS. Their direct involvement with the blood, bone marrow, and extramedullary sites complicates treatment management. However, recent therapeutic advancements offer hope for the long-term management of AL and MDS. This systematic review followed the guidelines put forth by Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) to assess recent developments and challenges in the treatment of AL (including acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL)) and MDS. Databases such as PubMed, Google Scholar, NCBI, Scopus, Blood Journal, Cochrane Library, and Leukemia Gene Atlas (LGA) were used to retrieve articles published from 2017 to 2024, with the last search conducted in August 2024. A total of 12 peer-reviewed studies were selected based on specific inclusion and exclusion criteria. These studies reveal advancements in the diagnosis, classification, and treatment of AL and MDS, including long-term disease-free survival, complete remissions, and improved patient outcomes in those over 75 years of age. Less toxic treatment methods, such as targeted therapies, immunotherapies, and bispecific T-cell engagers, are particularly beneficial for older adults with ALL. Significant progress has also been made in understanding the genetic mutations in AML, leading to more personalized therapies. In MDS, a combination of chemotherapy, immunosuppressive treatments, targeted therapies, and stem cell transplants has shown high efficacy. However, challenges remain, including high initial treatment costs, limited patient access, inadequate awareness, insufficient employee training, and the lack of accurate treatment models. Despite these hurdles, these advances provide promising options for improving the quality of life for patients with AL and MDS.
Collapse
Affiliation(s)
- Nawal Rafiq
- Accident and Emergency, Rehman Medical Institute, Peshawar, PAK
| | | | - Mashaal Sahibzada
- Medical High Dependency Unit, Northwest Teaching Hospital, Peshawar, PAK
| | | | | | - Najeeb Ullah
- Internal Medicine, Rehman Medical Institute, Peshawar, PAK
| | | | - Hira Khalil
- Internal Medicine, Rehman Medical Institute, Peshawar, PAK
| | - Umar Azam Ali
- Internal Medicine, Ayub Medical College, Abbottabad, PAK
| | | | | | - Ibrar Khaliq
- Internal Medicine, Services Hospital, Lahore, PAK
| |
Collapse
|
14
|
Korbecki J, Bosiacki M, Stasiak P, Snarski E, Brodowska A, Chlubek D, Baranowska-Bosiacka I. Clinical Aspects and Significance of β-Chemokines, γ-Chemokines, and δ-Chemokines in Molecular Cancer Processes in Acute Myeloid Leukemia (AML) and Myelodysplastic Neoplasms (MDS). Cancers (Basel) 2024; 16:3246. [PMID: 39409868 PMCID: PMC11476337 DOI: 10.3390/cancers16193246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/15/2024] [Accepted: 09/20/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND/OBJECTIVES Acute myeloid leukemia (AML) is a type of leukemia with a very poor prognosis. Consequently, this neoplasm is extensively researched to discover new therapeutic strategies. One area of investigation is the study of intracellular communication and the impact of the bone marrow microenvironment on AML cells, with chemokines being a key focus. The roles of β-chemokines, γ-chemokines, and δ-chemokines in AML processes have not yet been sufficiently characterized. METHODS This publication summarizes all available knowledge about these chemotactic cytokines in AML and myelodysplastic neoplasm (MDS) processes and presents potential therapeutic strategies to combat the disease. The significance of β-chemokines, γ-chemokines, and δ-chemokines is detailed, including CCL2 (MCP-1), CCL3 (MIP-1α), CCL5 (RANTES), CCL23, CCL28, and CX3CL1 (fractalkine). Additionally, the importance of atypical chemokine receptors in AML is discussed, specifically ACKR1, ACKR2, ACKR4, and CCRL2. RESULTS/CONCLUSIONS The focus is on the effects of these chemokines on AML cells, particularly their influence on proliferation and resistance to anti-leukemic drugs. Intercellular interactions with non-AML cells, such as mesenchymal stem cells (MSC), macrophages, and regulatory T cells (Treg), are also characterized. The clinical aspects of chemokines are thoroughly explained, including their effect on overall survival and the relationship between their blood levels and AML characteristics.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Anatomy and Histology, Collegium Medicum, University of Zielona Góra, Zyty 28, 65-046 Zielona Góra, Poland; (J.K.); (P.S.)
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (M.B.)
| | - Mateusz Bosiacki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (M.B.)
| | - Piotr Stasiak
- Department of Anatomy and Histology, Collegium Medicum, University of Zielona Góra, Zyty 28, 65-046 Zielona Góra, Poland; (J.K.); (P.S.)
| | - Emilian Snarski
- Institute of Medical Sciences, Collegium Medicum, University of Zielona Góra, Zyty 28 Str., 65-046 Zielona Góra, Poland;
| | - Agnieszka Brodowska
- Department of Gynecology, Endocrinology and Gynecological Oncology, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, 71-252 Szczecin, Poland;
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (M.B.)
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (M.B.)
| |
Collapse
|
15
|
Khattab S, El Sorady M, El-Ghandour A, Visani G, Piccaluga PP. Hematopoietic and leukemic stem cells homeostasis: the role of bone marrow niche. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:1027-1055. [PMID: 39351440 PMCID: PMC11438561 DOI: 10.37349/etat.2024.00262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 07/01/2024] [Indexed: 10/04/2024] Open
Abstract
The bone marrow microenvironment (BMM) has highly specialized anatomical characteristics that provide a sanctuary place for hematopoietic stem cells (HSCs) that allow appropriate proliferation, maintenance, and self-renewal capacity. Several cell types contribute to the constitution and function of the bone marrow niche. Interestingly, uncovering the secrets of BMM and its interaction with HSCs in health paved the road for research aiming at better understanding the concept of leukemic stem cells (LSCs) and their altered niche. In fact, they share many signals that are responsible for interactions between LSCs and the bone marrow niche, due to several biological similarities between LSCs and HSCs. On the other hand, LSCs differ from HSCs in their abnormal activation of important signaling pathways that regulate survival, proliferation, drug resistance, invasion, and spread. Targeting these altered niches can help in better treatment choices for hematological malignancies and bone marrow disorders in general and acute myeloid leukemia (AML) in particular. Moreover, targeting those niches may help in decreasing the emergence of drug resistance and lower the relapse rate. In this article, the authors reviewed the most recent literature on bone marrow niches and their relations with either normal HSCs and AML cells/LSC, by focusing on pathogenetic and therapeutic implications.
Collapse
Affiliation(s)
- Shaimaa Khattab
- Biobank of Research, IRCCS Azienda Ospedaliera-Universitaria di Bologna Policlinico di S. Orsola, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences, Bologna University School of Medicine, 40138 Bologna, Italy
- Medical Research Institute, Hematology department, Alexandria University, Alexandria 21561, Egypt
| | - Manal El Sorady
- Department of Internal Medicine, Faculty of Medicine, Alexandria University, Alexandria 5310002, Egypt
| | - Ashraf El-Ghandour
- Department of Internal Medicine, Faculty of Medicine, Alexandria University, Alexandria 5310002, Egypt
| | - Giuseppe Visani
- Hematology and Stem Cell Transplant Center, Azienda Ospedaliera Marche Nord, 61121 Pesaro, Italy
| | - Pier Paolo Piccaluga
- Biobank of Research, IRCCS Azienda Ospedaliera-Universitaria di Bologna Policlinico di S. Orsola, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences, Bologna University School of Medicine, 40138 Bologna, Italy
| |
Collapse
|
16
|
Olejarz W, Sadowski K, Szulczyk D, Basak G. Advancements in Personalized CAR-T Therapy: Comprehensive Overview of Biomarkers and Therapeutic Targets in Hematological Malignancies. Int J Mol Sci 2024; 25:7743. [PMID: 39062986 PMCID: PMC11276786 DOI: 10.3390/ijms25147743] [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: 06/30/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy is a novel anticancer therapy using autologous or allogeneic T-cells. To date, six CAR-T therapies for specific B-cell acute lymphoblastic leukemia (B-ALL), non-Hodgkin lymphomas (NHL), and multiple myeloma (MM) have been approved by the Food and Drug Administration (FDA). Significant barriers to the effectiveness of CAR-T therapy include cytokine release syndrome (CRS), neurotoxicity in the case of Allogeneic Stem Cell Transplantation (Allo-SCT) graft-versus-host-disease (GVHD), antigen escape, modest antitumor activity, restricted trafficking, limited persistence, the immunosuppressive microenvironment, and senescence and exhaustion of CAR-Ts. Furthermore, cancer drug resistance remains a major problem in clinical practice. CAR-T therapy, in combination with checkpoint blockades and bispecific T-cell engagers (BiTEs) or other drugs, appears to be an appealing anticancer strategy. Many of these agents have shown impressive results, combining efficacy with tolerability. Biomarkers like extracellular vesicles (EVs), cell-free DNA (cfDNA), circulating tumor (ctDNA) and miRNAs may play an important role in toxicity, relapse assessment, and efficacy prediction, and can be implicated in clinical applications of CAR-T therapy and in establishing safe and efficacious personalized medicine. However, further research is required to fully comprehend the particular side effects of immunomodulation, to ascertain the best order and combination of this medication with conventional chemotherapy and targeted therapies, and to find reliable predictive biomarkers.
Collapse
Affiliation(s)
- Wioletta Olejarz
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Karol Sadowski
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Daniel Szulczyk
- Chair and Department of Biochemistry, The Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Grzegorz Basak
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland;
| |
Collapse
|
17
|
Korbecki J, Bosiacki M, Szatkowska I, Kupnicka P, Chlubek D, Baranowska-Bosiacka I. The Clinical Significance and Involvement in Molecular Cancer Processes of Chemokine CXCL1 in Selected Tumors. Int J Mol Sci 2024; 25:4365. [PMID: 38673949 PMCID: PMC11050300 DOI: 10.3390/ijms25084365] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Chemokines play a key role in cancer processes, with CXCL1 being a well-studied example. Due to the lack of a complete summary of CXCL1's role in cancer in the literature, in this study, we examine the significance of CXCL1 in various cancers such as bladder, glioblastoma, hemangioendothelioma, leukemias, Kaposi's sarcoma, lung, osteosarcoma, renal, and skin cancers (malignant melanoma, basal cell carcinoma, and squamous cell carcinoma), along with thyroid cancer. We focus on understanding how CXCL1 is involved in the cancer processes of these specific types of tumors. We look at how CXCL1 affects cancer cells, including their proliferation, migration, EMT, and metastasis. We also explore how CXCL1 influences other cells connected to tumors, like promoting angiogenesis, recruiting neutrophils, and affecting immune cell functions. Additionally, we discuss the clinical aspects by exploring how CXCL1 levels relate to cancer staging, lymph node metastasis, patient outcomes, chemoresistance, and radioresistance.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (J.K.); (M.B.); (D.C.)
- Department of Anatomy and Histology, Collegium Medicum, University of Zielona Góra, Zyty 28, 65-046 Zielona Góra, Poland
| | - Mateusz Bosiacki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (J.K.); (M.B.); (D.C.)
| | - Iwona Szatkowska
- Department of Ruminants Science, Faculty of Biotechnology and Animal Husbandry, West Pomeranian University of Technology, Klemensa Janickiego 29 St., 71-270 Szczecin, Poland;
| | - Patrycja Kupnicka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (J.K.); (M.B.); (D.C.)
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (J.K.); (M.B.); (D.C.)
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (J.K.); (M.B.); (D.C.)
| |
Collapse
|
18
|
Sanz-Ortega L, Andersson A, Carlsten M. Harnessing upregulated E-selectin while enhancing SDF-1α sensing redirects infused NK cells to the AML-perturbed bone marrow. Leukemia 2024; 38:579-589. [PMID: 38182818 PMCID: PMC10912028 DOI: 10.1038/s41375-023-02126-1] [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: 07/27/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 01/07/2024]
Abstract
Increased bone marrow (BM) homing of NK cells is associated with positive outcome in patients with acute myeloid leukemia (AML) treated within adoptive NK cell transfer trials. While most efforts to further improve the efficacy focus on augmenting NK cell persistence and cytotoxicity, few address their ability to home to the tumor. Here, we decipher how AML growth alters the BM niche to impair NK cell infiltration and how insights can be utilized to resolve this issue. We show that AML development gradually impairs the BM homing capacity of infused NK cells, which was tightly linked to loss of SDF-1α in this environment. AML development also triggered up-regulation of E-selectin on BM endothelial cells. Given the poor E-selectin-binding capacity of NK cells, introduction of fucosyltransferase-7 (FUT7) to the NK cells per mRNA transfection resulted in potent E-selectin binding and stronger adhesion to E-selectin+ endothelial cells. Co-introduction of FUT7 and gain-of-function CXCR4 (CXCR4R334X) redirected NK cell homing to the BM of AML-bearing mice nearly to the levels in AML-free mice. This work shows how impaired NK cell homing caused by AML-induced microenvironmental changes can be overcome by genetic engineering. We speculate our insights can help further advance future NK cell immunotherapies.
Collapse
Affiliation(s)
- Laura Sanz-Ortega
- Center for Hematology and Regenerative Medicine, Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Agneta Andersson
- Center for Hematology and Regenerative Medicine, Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Mattias Carlsten
- Center for Hematology and Regenerative Medicine, Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden.
- Center for Cell Therapy and Allogeneic Stem Cell Transplantation, Karolinska Comprehensive Cancer Center, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
19
|
Wu Y, Li Y, Gao Y, Zhang P, Jing Q, Zhang Y, Jin W, Wang Y, Du J, Wu G. Immunotherapies of acute myeloid leukemia: Rationale, clinical evidence and perspective. Biomed Pharmacother 2024; 171:116132. [PMID: 38198961 DOI: 10.1016/j.biopha.2024.116132] [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: 09/30/2023] [Revised: 12/28/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024] Open
Abstract
Acute myeloid leukemia (AML) is a prevalent hematological malignancy that exhibits a wide array of molecular abnormalities. Although traditional treatment modalities such as chemotherapy and allogeneic stem cell transplantation (HSCT) have become standard therapeutic approaches, a considerable number of patients continue to face relapse and encounter a bleak prognosis. The emergence of immune escape, immunosuppression, minimal residual disease (MRD), and other contributing factors collectively contribute to this challenge. Recent research has increasingly highlighted the notable distinctions between AML tumor microenvironments and those of healthy individuals. In order to investigate the potential therapeutic mechanisms, this study examines the intricate transformations occurring between leukemic cells and their surrounding cells within the tumor microenvironment (TME) of AML. This review classifies immunotherapies into four distinct categories: cancer vaccines, immune checkpoint inhibitors (ICIs), antibody-based immunotherapies, and adoptive T-cell therapies. The results of numerous clinical trials strongly indicate that the identification of optimal combinations of novel agents, either in conjunction with each other or with chemotherapy, represents a crucial advancement in this field. In this review, we aim to explore the current and emerging immunotherapeutic methodologies applicable to AML patients, identify promising targets, and emphasize the crucial requirement to augment patient outcomes. The application of these strategies presents substantial therapeutic prospects within the realm of precision medicine for AML, encompassing the potential to ameliorate patient outcomes.
Collapse
Affiliation(s)
- Yunyi Wu
- Department of Central Laboratory, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China; Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yanchun Li
- Department of Central Laboratory, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Yan Gao
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ping Zhang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qiangan Jing
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yinhao Zhang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Weidong Jin
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ying Wang
- Department of Central Laboratory, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China.
| | - Jing Du
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Gongqiang Wu
- Department of Hematology, Dongyang Hospitai Affiliated to Wenzhou Medical University, Dongyang People's Hospital, Dongyang, Zhejiang, China.
| |
Collapse
|
20
|
Tomasoni C, Arsuffi C, Donsante S, Corsi A, Riminucci M, Biondi A, Pievani A, Serafini M. AML alters bone marrow stromal cell osteogenic commitment via Notch signaling. Front Immunol 2023; 14:1320497. [PMID: 38111584 PMCID: PMC10725948 DOI: 10.3389/fimmu.2023.1320497] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 11/20/2023] [Indexed: 12/20/2023] Open
Abstract
Introduction Acute myeloid leukemia (AML) is a highly heterogeneous malignancy caused by various genetic alterations and characterized by the accumulation of immature myeloid blasts in the bone marrow (BM). This abnormal growth of AML cells disrupts normal hematopoiesis and alters the BM microenvironment components, establishing a niche supportive of leukemogenesis. Bone marrow stromal cells (BMSCs) play a pivotal role in giving rise to essential elements of the BM niche, including adipocytes and osteogenic cells. Animal models have shown that the BM microenvironment is significantly remodeled by AML cells, which skew BMSCs toward an ineffective osteogenic differentiation with an accumulation of osteoprogenitors. However, little is known about the mechanisms by which AML cells affect osteogenesis. Methods We studied the effect of AML cells on the osteogenic commitment of normal BMSCs, using a 2D co-culture system. Results We found that AML cell lines and primary blasts, but not normal hematopoietic CD34+ cells, induced in BMSCs an ineffective osteogenic commitment, with an increase of the early-osteogenic marker tissue non-specific alkaline phosphatase (TNAP) in the absence of the late-osteogenic gene up-regulation. Moreover, the direct interaction of AML cells and BMSCs was indispensable in influencing osteogenic differentiation. Mechanistic studies identified a role for AML-mediated Notch activation in BMSCs contributing to their ineffective osteogenic commitment. Inhibition of Notch using a γ-secretase inhibitor strongly influenced Notch signaling in BMSCs and abrogated the AML-induced TNAP up-regulation. Discussion Together, our data support the hypothesis that AML infiltration produces a leukemia-supportive pre-osteoblast-rich niche in the BM, which can be partially ascribed to AML-induced activation of Notch signaling in BMSCs.
Collapse
Affiliation(s)
- Chiara Tomasoni
- Tettamanti Center, Fondazione Istituto Ricovero e Cura a Carattere Scientifico (IRCCS) San Gerardo dei Tintori, Monza, Italy
| | - Corinne Arsuffi
- Tettamanti Center, Fondazione Istituto Ricovero e Cura a Carattere Scientifico (IRCCS) San Gerardo dei Tintori, Monza, Italy
| | - Samantha Donsante
- Tettamanti Center, Fondazione Istituto Ricovero e Cura a Carattere Scientifico (IRCCS) San Gerardo dei Tintori, Monza, Italy
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Alessandro Corsi
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Mara Riminucci
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Andrea Biondi
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Pediatrics, Fondazione Istituto Ricovero e Cura a Carattere Scientifico (IRCCS) San Gerardo dei Tintori, Monza, Italy
| | - Alice Pievani
- Tettamanti Center, Fondazione Istituto Ricovero e Cura a Carattere Scientifico (IRCCS) San Gerardo dei Tintori, Monza, Italy
| | - Marta Serafini
- Tettamanti Center, Fondazione Istituto Ricovero e Cura a Carattere Scientifico (IRCCS) San Gerardo dei Tintori, Monza, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
21
|
Alhattab DM, Isaioglou I, Alshehri S, Khan ZN, Susapto HH, Li Y, Marghani Y, Alghuneim AA, Díaz-Rúa R, Abdelrahman S, Al-Bihani S, Ahmed F, Felimban RI, Alkhatabi H, Alserihi R, Abedalthagafi M, AlFadel A, Awidi A, Chaudhary AG, Merzaban J, Hauser CAE. Fabrication of a three-dimensional bone marrow niche-like acute myeloid Leukemia disease model by an automated and controlled process using a robotic multicellular bioprinting system. Biomater Res 2023; 27:111. [PMID: 37932837 PMCID: PMC10626721 DOI: 10.1186/s40824-023-00457-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/29/2023] [Indexed: 11/08/2023] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a hematological malignancy that remains a therapeutic challenge due to the high incidence of disease relapse. To better understand resistance mechanisms and identify novel therapies, robust preclinical models mimicking the bone marrow (BM) microenvironment are needed. This study aimed to achieve an automated fabrication process of a three-dimensional (3D) AML disease model that recapitulates the 3D spatial structure of the BM microenvironment and applies to drug screening and investigational studies. METHODS To build this model, we investigated a unique class of tetramer peptides with an innate ability to self-assemble into stable hydrogel. An automated robotic bioprinting process was established to fabricate a 3D BM (niche-like) multicellular AML disease model comprised of leukemia cells and the BM's stromal and endothelial cellular fractions. In addition, monoculture and dual-culture models were also fabricated. Leukemia cell compatibility, functionalities (in vitro and in vivo), and drug assessment studies using our model were performed. In addition, RNAseq and gene expression analysis using TaqMan arrays were also performed on 3D cultured stromal cells and primary leukemia cells. RESULTS The selected peptide hydrogel formed a highly porous network of nanofibers with mechanical properties similar to the BM extracellular matrix. The robotic bioprinter and the novel quadruple coaxial nozzle enabled the automated fabrication of a 3D BM niche-like AML disease model with controlled deposition of multiple cell types into the model. This model supported the viability and growth of primary leukemic, endothelial, and stromal cells and recapitulated cell-cell and cell-ECM interactions. In addition, AML cells in our model possessed quiescent characteristics with improved chemoresistance attributes, resembling more the native conditions as indicated by our in vivo results. Moreover, the whole transcriptome data demonstrated the effect of 3D culture on enhancing BM niche cell characteristics. We identified molecular pathways upregulated in AML cells in our 3D model that might contribute to AML drug resistance and disease relapse. CONCLUSIONS Our results demonstrate the importance of developing 3D biomimicry models that closely recapitulate the in vivo conditions to gain deeper insights into drug resistance mechanisms and novel therapy development. These models can also improve personalized medicine by testing patient-specific treatments.
Collapse
Affiliation(s)
- Dana M Alhattab
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- KAUST Smart Health Initiative (KSHI), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Ioannis Isaioglou
- Cell Migration and Signaling Laboratory, Bioscience Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Salwa Alshehri
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Zainab N Khan
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- Red Sea Research Center (RSRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Hepi H Susapto
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Yanyan Li
- Cell Migration and Signaling Laboratory, Bioscience Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Yara Marghani
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Arwa A Alghuneim
- Cell Migration and Signaling Laboratory, Bioscience Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Rubén Díaz-Rúa
- Core Laboratories, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Sherin Abdelrahman
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Shuroug Al-Bihani
- Core Laboratories, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Farid Ahmed
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Innovation in Personalized Medicine (CIPM), King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Raed I Felimban
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Innovation in Personalized Medicine (CIPM), King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Heba Alkhatabi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Innovation in Personalized Medicine (CIPM), King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Hematology Research Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Raed Alserihi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Innovation in Personalized Medicine (CIPM), King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Malak Abedalthagafi
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, USA
| | - AlShaibani AlFadel
- Division of Hematology, Stem Cell Transplantation & Cellular Therapy, Oncology Center, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Abdalla Awidi
- Cell Therapy Center, The University of Jordan, Amman, Jordan
- Medical School, The University of Jordan, Amman, Jordan
- Jordan University Hospital, Amman, Jordan
| | - Adeel Gulzar Chaudhary
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Innovation in Personalized Medicine (CIPM), King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Jasmeen Merzaban
- Cell Migration and Signaling Laboratory, Bioscience Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Charlotte A E Hauser
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.
- KAUST Smart Health Initiative (KSHI), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.
- Red Sea Research Center (RSRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.
| |
Collapse
|
22
|
Molica M, Perrone S, Andriola C, Rossi M. Immunotherapy with Monoclonal Antibodies for Acute Myeloid Leukemia: A Work in Progress. Cancers (Basel) 2023; 15:5060. [PMID: 37894427 PMCID: PMC10605302 DOI: 10.3390/cancers15205060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/12/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
In the last few years, molecularly targeted agents and immune-based treatments (ITs) have significantly changed the landscape of anti-cancer therapy. Indeed, ITs have been proven to be very effective when used against metastatic solid tumors, for which outcomes are extremely poor when using standard approaches. Such a scenario has only been partially reproduced in hematologic malignancies. In the context of acute myeloid leukemia (AML), as innovative drugs are eagerly awaited in the relapsed/refractory setting, different ITs have been explored, but the results are still unsatisfactory. In this work, we will discuss the most important clinical studies to date that adopt ITs in AML, providing the basis to understand how this approach, although still in its infancy, may represent a promising therapeutic tool for the future treatment of AML patients.
Collapse
Affiliation(s)
- Matteo Molica
- Department of Hematology-Oncology, Azienda Universitaria Ospedaliera Renato Dulbecco, 88100 Catanzaro, Italy;
| | - Salvatore Perrone
- Department of Hematology, Polo Universitario Pontino, S.M. Goretti Hospital, 04100 Latina, Italy;
| | - Costanza Andriola
- Hematology, Department of Translational and Precision Medicine, Sapienza University, 00100 Rome, Italy;
| | - Marco Rossi
- Department of Hematology-Oncology, Azienda Universitaria Ospedaliera Renato Dulbecco, 88100 Catanzaro, Italy;
| |
Collapse
|
23
|
Peroni E, Randi ML, Rosato A, Cagnin S. Acute myeloid leukemia: from NGS, through scRNA-seq, to CAR-T. dissect cancer heterogeneity and tailor the treatment. J Exp Clin Cancer Res 2023; 42:259. [PMID: 37803464 PMCID: PMC10557350 DOI: 10.1186/s13046-023-02841-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 09/25/2023] [Indexed: 10/08/2023] Open
Abstract
Acute myeloid leukemia (AML) is a malignant blood cancer with marked cellular heterogeneity due to altered maturation and differentiation of myeloid blasts, the possible causes of which are transcriptional or epigenetic alterations, impaired apoptosis, and excessive cell proliferation. This neoplasm has a high rate of resistance to anticancer therapies and thus a high risk of relapse and mortality because of both the biological diversity of the patient and intratumoral heterogeneity due to the acquisition of new somatic changes. For more than 40 years, the old gold standard "one size fits all" treatment approach included intensive chemotherapy treatment with anthracyclines and cytarabine.The manuscript first traces the evolution of the understanding of the pathology from the 1970s to the present. The enormous strides made in its categorization prove to be crucial for risk stratification, enabling an increasingly personalized diagnosis and treatment approach.Subsequently, we highlight how, over the past 15 years, technological advances enabling single cell RNA sequencing and T-cell modification based on the genomic tools are affecting the classification and treatment of AML. At the dawn of the new millennium, the advent of high-throughput next-generation sequencing technologies has enabled the profiling of patients evidencing different facets of the same disease, stratifying risk, and identifying new possible therapeutic targets that have subsequently been validated. Currently, the possibility of investigating tumor heterogeneity at the single cell level, profiling the tumor at the time of diagnosis or after treatments exist. This would allow the identification of underrepresented cellular subclones or clones resistant to therapeutic approaches and thus responsible for post-treatment relapse that would otherwise be difficult to detect with bulk investigations on the tumor biopsy. Single-cell investigation will then allow even greater personalization of therapy to the genetic and transcriptional profile of the tumor, saving valuable time and dangerous side effects. The era of personalized medicine will take a huge step forward through the disclosure of each individual piece of the complex puzzle that is cancer pathology, to implement a "tailored" therapeutic approach based also on engineered CAR-T cells.
Collapse
Affiliation(s)
- Edoardo Peroni
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology, IOV-IRCCS, Padova, 35128, Italy.
| | - Maria Luigia Randi
- First Medical Clinic, Department of Medicine-DIMED, University of Padua, Padua, Italy
| | - Antonio Rosato
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology, IOV-IRCCS, Padova, 35128, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Stefano Cagnin
- Department of Biology, University of Padova, Padova, 35131, Italy
- CIR-Myo Myology Center, University of Padova, Padova, 35131, Italy
| |
Collapse
|
24
|
Humphries S, Bond DR, Germon ZP, Keely S, Enjeti AK, Dun MD, Lee HJ. Crosstalk between DNA methylation and hypoxia in acute myeloid leukaemia. Clin Epigenetics 2023; 15:150. [PMID: 37705055 PMCID: PMC10500762 DOI: 10.1186/s13148-023-01566-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/08/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND Acute myeloid leukaemia (AML) is a deadly disease characterised by the uncontrolled proliferation of immature myeloid cells within the bone marrow. Altered regulation of DNA methylation is an important epigenetic driver of AML, where the hypoxic bone marrow microenvironment can help facilitate leukaemogenesis. Thus, interactions between epigenetic regulation and hypoxia signalling will have important implications for AML development and treatment. MAIN BODY This review summarises the importance of DNA methylation and the hypoxic bone marrow microenvironment in the development, progression, and treatment of AML. Here, we focus on the role hypoxia plays on signalling and the subsequent regulation of DNA methylation. Hypoxia is likely to influence DNA methylation through altered metabolic pathways, transcriptional control of epigenetic regulators, and direct effects on the enzymatic activity of epigenetic modifiers. DNA methylation may also prevent activation of hypoxia-responsive genes, demonstrating bidirectional crosstalk between epigenetic regulation and the hypoxic microenvironment. Finally, we consider the clinical implications of these interactions, suggesting that reduced cell cycling within the hypoxic bone marrow may decrease the efficacy of hypomethylating agents. CONCLUSION Hypoxia is likely to influence AML progression through complex interactions with DNA methylation, where the therapeutic efficacy of hypomethylating agents may be limited within the hypoxic bone marrow. To achieve optimal outcomes for AML patients, future studies should therefore consider co-treatments that can promote cycling of AML cells within the bone marrow or encourage their dissociation from the bone marrow.
Collapse
Affiliation(s)
- Sam Humphries
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2308, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Danielle R Bond
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2308, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Zacary P Germon
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2308, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Simon Keely
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2308, Australia
- Immune Health Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Anoop K Enjeti
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
- Department of Haematology, Calvary Mater Hospital, Waratah, NSW, 2298, Australia
- New South Wales Health Pathology, John Hunter Hospital, New Lambton Heights, NSW, 2305, Australia
| | - Matthew D Dun
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2308, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Heather J Lee
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2308, Australia.
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia.
| |
Collapse
|
25
|
Madaci L, Gard C, Nin S, Venton G, Rihet P, Puthier D, Loriod B, Costello R. The Contribution of Multiplexing Single Cell RNA Sequencing in Acute Myeloid Leukemia. Diseases 2023; 11:96. [PMID: 37489448 PMCID: PMC10366847 DOI: 10.3390/diseases11030096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/26/2023] Open
Abstract
Decades ago, the treatment for acute myeloid leukemia relied on cytarabine and anthracycline. However, advancements in medical research have introduced targeted therapies, initially employing monoclonal antibodies such as ant-CD52 and anti-CD123, and subsequently utilizing specific inhibitors that target molecular mutations like anti-IDH1, IDH2, or FLT3. The challenge lies in determining the role of these therapeutic options, considering the inherent tumor heterogeneity associated with leukemia diagnosis and the clonal drift that this type of tumor can undergo. Targeted drugs necessitate an examination of various therapeutic targets at the individual cell level rather than assessing the entire population. It is crucial to differentiate between the prognostic value and therapeutic potential of a specific molecular target, depending on whether it is found in a terminally differentiated cell with limited proliferative potential or a stem cell with robust capabilities for both proliferation and self-renewal. However, this cell-by-cell analysis is accompanied by several challenges. Firstly, the scientific aspect poses difficulties in comparing different single cell analysis experiments despite efforts to standardize the results through various techniques. Secondly, there are practical obstacles as each individual cell experiment incurs significant financial costs and consumes a substantial amount of time. A viable solution lies in the ability to process multiple samples simultaneously, which is a distinctive feature of the cell hashing technique. In this study, we demonstrate the applicability of the cell hashing technique for analyzing acute myeloid leukemia cells. By comparing it to standard single cell analysis, we establish a strong correlation in various parameters such as quality control, gene expression, and the analysis of leukemic blast markers in patients. Consequently, this technique holds the potential to become an integral part of the biological assessment of acute myeloid leukemia, contributing to the personalized and optimized management of the disease, particularly in the context of employing targeted therapies.
Collapse
Affiliation(s)
- Lamia Madaci
- TAGC, INSERM, UMR1090, Aix Marseille University, Parc Scientifique de Luminy, 13009 Marseille, France
| | - Charlyne Gard
- TAGC, INSERM, UMR1090, Aix Marseille University, Parc Scientifique de Luminy, 13009 Marseille, France
| | - Sébastien Nin
- TAGC, INSERM, UMR1090, Aix Marseille University, Parc Scientifique de Luminy, 13009 Marseille, France
| | - Geoffroy Venton
- TAGC, INSERM, UMR1090, Aix Marseille University, Parc Scientifique de Luminy, 13009 Marseille, France
- Hematology and Cellular Therapy Department, Conception Hospital, 13005 Marseille, France
| | - Pascal Rihet
- TAGC, INSERM, UMR1090, Aix Marseille University, Parc Scientifique de Luminy, 13009 Marseille, France
| | - Denis Puthier
- TAGC, INSERM, UMR1090, Aix Marseille University, Parc Scientifique de Luminy, 13009 Marseille, France
| | - Béatrice Loriod
- TAGC, INSERM, UMR1090, Aix Marseille University, Parc Scientifique de Luminy, 13009 Marseille, France
| | - Régis Costello
- TAGC, INSERM, UMR1090, Aix Marseille University, Parc Scientifique de Luminy, 13009 Marseille, France
- Hematology and Cellular Therapy Department, Conception Hospital, 13005 Marseille, France
| |
Collapse
|
26
|
Alberti G, Arsuffi C, Pievani A, Salerno D, Mantegazza F, Dazzi F, Biondi A, Tettamanti S, Serafini M. Engineering tandem CD33xCD146 CAR CIK (cytokine-induced killer) cells to target the acute myeloid leukemia niche. Front Immunol 2023; 14:1192333. [PMID: 37304257 PMCID: PMC10247966 DOI: 10.3389/fimmu.2023.1192333] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/12/2023] [Indexed: 06/13/2023] Open
Abstract
In acute myeloid leukemia (AML), malignant stem cells hijack the normal bone marrow niche where they are largely protected from the current therapeutic approaches. Thus, eradicating these progenitors is the ultimate challenge in the treatment of this disease. Specifically, the development of chimeric antigen receptors (CARs) against distinct mesenchymal stromal cell subpopulations involved in the maintenance of leukemic stem cells within the malignant bone marrow microenvironment could represent a new strategy to improve CAR T-cell therapy efficacy, which is still unsuccessful in AML. As a proof of concept, we generated a novel prototype of Tandem CAR, with one specificity directed against the leukemic cell marker CD33 and the other against the mesenchymal stromal cell marker CD146, demonstrating its capability of simultaneously targeting two different cell types in a 2D co-culture system. Interestingly, we could also observe an in vitro inhibition of CAR T cell functionality mediated by stromal cells, particularly in later effector functions, such as reduction of interferon-gamma and interleukin-2 release and impaired proliferation of the CAR+ effector Cytokine-Induced Killer (CIK) cells. Taken together, these data demonstrate the feasibility of a dual targeting model against two molecules, which are expressed on two different target cells, but also highlight the immunomodulatory effect on CAR CIK cells exerted by stromal cells, confirming that the niche could be an obstacle to the efficacy of CAR T cells. This aspect should be considered in the development of novel CAR T cell approaches directed against the AML bone marrow niche.
Collapse
Affiliation(s)
- Gaia Alberti
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Corinne Arsuffi
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Alice Pievani
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Domenico Salerno
- School of Medicine and Surgery, BioNanoMedicine Center NANOMIB, Universita di Milano-Bicocca, Vedano al Lambro (MB), Italy
| | - Francesco Mantegazza
- School of Medicine and Surgery, BioNanoMedicine Center NANOMIB, Universita di Milano-Bicocca, Vedano al Lambro (MB), Italy
| | - Francesco Dazzi
- School of Cardiovascular Sciences, King’s College London, London, United Kingdom
| | - Andrea Biondi
- Dipartimento di Medicina e Chirurgia, Università degli Studi Milano-Bicocca, Monza (MB), Italy
- Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Sarah Tettamanti
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Marta Serafini
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| |
Collapse
|
27
|
Kotsiafti A, Giannakas K, Christoforou P, Liapis K. Progress toward Better Treatment of Therapy-Related AML. Cancers (Basel) 2023; 15:cancers15061658. [PMID: 36980546 PMCID: PMC10046015 DOI: 10.3390/cancers15061658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/30/2023] Open
Abstract
Therapy-related acute myeloid leukemia (t-AML) comprises 10-20% of all newly diagnosed cases of AML and is related to previous use of chemotherapy or ionizing radiotherapy for an unrelated malignant non-myeloid disorder or autoimmune disease. Classic examples include alkylating agents and topoisomerase II inhibitors, whereas newer targeted therapies such as poly (adenosine diphosphate-ribose) polymerase (PARP) inhibitors have emerged as causative agents. Typically, t-AML is characterized by adverse karyotypic abnormalities and molecular lesions that confer a poor prognosis. Nevertheless, there are also cases of t-AML without poor-risk features. The management of these patients remains controversial. We describe the causes and pathophysiology of t-AML, putting emphasis on its mutational heterogeneity, and present recent advances in its treatment including CPX-351, hypomethylating agent plus venetoclax combination, and novel, molecularly targeted agents that promise to improve the cure rates. Evidence supporting personalized medicine for patients with t-AML is presented, as well as the authors' clinical recommendations.
Collapse
Affiliation(s)
| | | | - Panagiotis Christoforou
- Pathophysiology Department, National and Kapodistrian University of Athens, 157 72 Athens, Greece
| | - Konstantinos Liapis
- Dragana Campus, Democritus University of Thrace Medical School, 681 00 Alexandroupolis, Greece
| |
Collapse
|
28
|
Nath P, Modak S, Aktar T, Maiti S, Ghosh A, Singh R, Debnath M, Saha B, Maiti D. Olive leaves extract alleviates inflammation and modifies the intrinsic apoptotic signal in the leukemic bone marrow. Front Immunol 2023; 13:1054186. [PMID: 36741365 PMCID: PMC9894250 DOI: 10.3389/fimmu.2022.1054186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/19/2022] [Indexed: 01/20/2023] Open
Abstract
Introduction Current anti-leukemic chemotherapies with multiple targets suffer from side effects. Synthetic drugs with huge off-target effects are detrimental to leukemic patients. Therefore, natural plant-based products are being increasingly tested for new anti-leukemic therapy with fewer or no side effects. Herein, we report the effect of ethanolic olive leaves extract (EOLE) on the K562 cell line and on the bone marrow (BM) of N-ethyl-N-nitrosourea (ENU)-induced leukemic mice. Methods Using standard methodologies, we assessed viability, chromatin condensation, and induction of apoptosis in EOLE-treated K562 cells in-vitro. The anti-leukemic activity of EOLE was assayed by measuring ROS, levels of various cytokines, expression of iNOS and COX-2 gene, and changes in the level of important apoptosis regulatory and cell signaling proteins in-vivo. Result K562 cells underwent apoptotic induction after exposure to EOLE. In the BM of leukemic mice, EOLE therapy decreased the number of blast cells, ROS generation, and expression of NF-κB and ERK1/2. IL-6, IL-1β, TNF-α, iNOS, and COX-2 were among the inflammatory molecules that were down-regulated by EOLE therapy. Additionally, it decreased the expression of anti-apoptotic proteins BCL2A1, BCL-xL, and MCL-1 in the BM of leukemic mice. Discussion Chronic inflammation and anomalous apoptotic mechanism both critically contribute to the malignant transformation of cells. Inflammation in the tumor microenvironment promotes the growth, survival, and migration of cancer cells, accelerating the disease. The current investigation showed that EOLE treatment reduces inflammation and alters the expression of apoptosis regulatory protein in the BM of leukemic mice, which may halt the progression of the disease.
Collapse
Affiliation(s)
- Priyatosh Nath
- Immunology Microbiology Laboratory, Department of Human Physiology, Tripura University, Agartala, Tripura, India
| | - Snehashish Modak
- Immunology Microbiology Laboratory, Department of Human Physiology, Tripura University, Agartala, Tripura, India
| | - Tamanna Aktar
- Immunology Microbiology Laboratory, Department of Human Physiology, Tripura University, Agartala, Tripura, India
| | - Sharanya Maiti
- Delhi Public School Megacity, Kolkata, West Bengal, India
| | - Anisha Ghosh
- Delhi Public School Megacity, Kolkata, West Bengal, India
| | - Riddha Singh
- Hariyana Vidyamandir, Kolkata, West Bengal, India
| | - Mousumi Debnath
- Department of Biosciences, Manipal University Jaipur, Jaipur, Rajasthan, India
| | - Bhaskar Saha
- National Centre for Cell Science, Pune, Maharashtra, India
| | - Debasish Maiti
- Immunology Microbiology Laboratory, Department of Human Physiology, Tripura University, Agartala, Tripura, India,*Correspondence: Debasish Maiti, ;
| |
Collapse
|
29
|
Ureña-Bailén G, Dobrowolski JM, Hou Y, Dirlam A, Roig-Merino A, Schleicher S, Atar D, Seitz C, Feucht J, Antony JS, Mohammadian Gol T, Handgretinger R, Mezger M. Preclinical Evaluation of CRISPR-Edited CAR-NK-92 Cells for Off-the-Shelf Treatment of AML and B-ALL. Int J Mol Sci 2022; 23:12828. [PMID: 36361619 PMCID: PMC9655234 DOI: 10.3390/ijms232112828] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/13/2022] [Accepted: 10/21/2022] [Indexed: 08/10/2023] Open
Abstract
Acute myeloid leukemia (AML) and B-cell acute lymphocytic leukemia (B-ALL) are severe blood malignancies affecting both adults and children. Chimeric antigen receptor (CAR)-based immunotherapies have proven highly efficacious in the treatment of leukemia. However, the challenge of the immune escape of cancer cells remains. The development of more affordable and ready-to-use therapies is essential in view of the costly and time-consuming preparation of primary cell-based treatments. In order to promote the antitumor function against AML and B-ALL, we transduced NK-92 cells with CD276-CAR or CD19-CAR constructs. We also attempted to enhance cytotoxicity by a gene knockout of three different inhibitory checkpoints in NK cell function (CBLB, NKG2A, TIGIT) with CRISPR-Cas9 technology. The antileukemic activity of the generated cell lines was tested with calcein and luciferase-based cytotoxicity assays in various leukemia cell lines. Both CAR-NK-92 exhibited targeted cytotoxicity and a significant boost in antileukemic function in comparison to parental NK-92. CRISPR-Cas9 knock-outs did not improve B-ALL cytotoxicity. However, triple knock-out CD276-CAR-NK-92 cells, as well as CBLB or TIGIT knock-out NK-92 cells, showed significantly enhanced cytotoxicity against U-937 or U-937 CD19/tag AML cell lines. These results indicate that the CD19-CAR and CD276-CAR-NK-92 cell lines' cytotoxic performance is suitable for leukemia killing, making them promising off-the-shelf therapeutic candidates. The knock-out of CBLB and TIGIT in NK-92 and CD276-CAR-NK-92 should be further investigated for the treatment of AML.
Collapse
MESH Headings
- Humans
- Antigens, CD19
- B7 Antigens/metabolism
- Cell Line, Tumor
- Cytotoxicity, Immunologic
- Immunotherapy, Adoptive/methods
- Killer Cells, Natural
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/metabolism
- Lymphoma, B-Cell
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism
- Receptors, Chimeric Antigen
Collapse
Affiliation(s)
- Guillermo Ureña-Bailén
- Department of Hematology and Oncology, Children’s Hospital, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Jérôme-Maurice Dobrowolski
- Department of Hematology and Oncology, Children’s Hospital, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Yujuan Hou
- Department of Hematology and Oncology, Children’s Hospital, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Alicia Dirlam
- Department of Hematology and Oncology, Children’s Hospital, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | | | - Sabine Schleicher
- Department of Hematology and Oncology, Children’s Hospital, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Daniel Atar
- Department of Hematology and Oncology, Children’s Hospital, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Christian Seitz
- Department of Hematology and Oncology, Children’s Hospital, University Hospital Tuebingen, 72076 Tuebingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, 72074 Tuebingen, Germany
| | - Judith Feucht
- Department of Hematology and Oncology, Children’s Hospital, University Hospital Tuebingen, 72076 Tuebingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, 72074 Tuebingen, Germany
| | - Justin S. Antony
- Department of Hematology and Oncology, Children’s Hospital, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Tahereh Mohammadian Gol
- Department of Hematology and Oncology, Children’s Hospital, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Rupert Handgretinger
- Department of Hematology and Oncology, Children’s Hospital, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Markus Mezger
- Department of Hematology and Oncology, Children’s Hospital, University Hospital Tuebingen, 72076 Tuebingen, Germany
| |
Collapse
|
30
|
Luciano M, Krenn PW, Horejs-Hoeck J. The cytokine network in acute myeloid leukemia. Front Immunol 2022; 13:1000996. [PMID: 36248849 PMCID: PMC9554002 DOI: 10.3389/fimmu.2022.1000996] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
Acute myeloid leukemia (AML) is a highly heterogeneous malignancy of the blood and bone marrow, characterized by clonal expansion of myeloid stem and progenitor cells and rapid disease progression. Chemotherapy has been the first-line treatment for AML for more than 30 years. Application of recent high-throughput next-generation sequencing technologies has revealed significant molecular heterogeneity to AML, which in turn has motivated efforts to develop new, targeted therapies. However, due to the high complexity of this disease, including multiple driver mutations and the coexistence of multiple competing tumorigenic clones, the successful incorporation of these new agents into clinical practice remains challenging. These continuing difficulties call for the identification of innovative therapeutic approaches that are effective for a larger cohort of AML patients. Recent studies suggest that chronic immune stimulation and aberrant cytokine signaling act as triggers for AML initiation and progression, facets of the disease which might be exploited as promising targets in AML treatment. However, despite the greater appreciation of cytokine profiles in AML, the exact functions of cytokines in AML pathogenesis are not fully understood. Therefore, unravelling the molecular basis of the complex cytokine networks in AML is a prerequisite to develop new therapeutic alternatives based on targeting cytokines and their receptors.
Collapse
Affiliation(s)
- Michela Luciano
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Peter W. Krenn
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Jutta Horejs-Hoeck
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| |
Collapse
|
31
|
Zhang X, Zhu L, Zhang H, Chen S, Xiao Y. CAR-T Cell Therapy in Hematological Malignancies: Current Opportunities and Challenges. Front Immunol 2022; 13:927153. [PMID: 35757715 PMCID: PMC9226391 DOI: 10.3389/fimmu.2022.927153] [Citation(s) in RCA: 155] [Impact Index Per Article: 51.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 05/16/2022] [Indexed: 12/13/2022] Open
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy represents a major breakthrough in cancer treatment, and it has achieved unprecedented success in hematological malignancies, especially in relapsed/refractory (R/R) B cell malignancies. At present, CD19 and BCMA are the most common targets in CAR-T cell therapy, and numerous novel therapeutic targets are being explored. However, the adverse events related to CAR-T cell therapy might be serious or even life-threatening, such as cytokine release syndrome (CRS), CAR-T-cell-related encephalopathy syndrome (CRES), infections, cytopenia, and CRS-related coagulopathy. In addition, due to antigen escape, the limited CAR-T cell persistence, and immunosuppressive tumor microenvironment, a considerable proportion of patients relapse after CAR-T cell therapy. Thus, in this review, we focus on the progress and challenges of CAR-T cell therapy in hematological malignancies, such as attractive therapeutic targets, CAR-T related toxicities, and resistance to CAR-T cell therapy, and provide some practical recommendations.
Collapse
Affiliation(s)
- Xiaomin Zhang
- Department of Hematology, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Institute of Clinical Medicine College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lingling Zhu
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Hui Zhang
- School of Medicine, Jishou University, Jishou, China
| | - Shanshan Chen
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yang Xiao
- Institute of Clinical Medicine College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Hematology, Shenzhen Qianhai Shekou Pilot Free Trade Zone Hospital, Shenzhen, China
| |
Collapse
|
32
|
Reikvam H, Hatfield KJ, Wendelbo Ø, Lindås R, Lassalle P, Bruserud Ø. Endocan in Acute Leukemia: Current Knowledge and Future Perspectives. Biomolecules 2022; 12:biom12040492. [PMID: 35454082 PMCID: PMC9027427 DOI: 10.3390/biom12040492] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/21/2022] [Accepted: 03/21/2022] [Indexed: 11/16/2022] Open
Abstract
Endocan is a soluble dermatan sulfate proteoglycan expressed by endothelial cells and detected in serum/plasma. Its expression is increased in tumors/tumor vessels in several human malignancies, and high expression (high serum/plasma levels or tumor levels) has an adverse prognostic impact in several malignancies. The p14 endocan degradation product can also be detected in serum/plasma, but previous clinical studies as well as previously unpublished results presented in this review suggest that endocan and p14 endocan fragment levels reflect different biological characteristics, and the endocan levels seem to reflect the disease heterogeneity in acute leukemia better than the p14 fragment levels. Furthermore, decreased systemic endocan levels in previously immunocompetent sepsis patients are associated with later severe respiratory complications, but it is not known whether this is true also for immunocompromised acute leukemia patients. Finally, endocan is associated with increased early nonrelapse mortality in (acute leukemia) patients receiving allogeneic stem cell transplantation, and this adverse prognostic impact seems to be independent of the adverse impact of excessive fluid overload. Systemic endocan levels may also become important to predict cytokine release syndrome after immunotherapy/haploidentical transplantation, and in the long-term follow-up of acute leukemia survivors with regard to cardiovascular risk. Therapeutic targeting of endocan is now possible, and the possible role of endocan in acute leukemia should be further investigated to clarify whether the therapeutic strategy should also be considered.
Collapse
Affiliation(s)
- Håkon Reikvam
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway;
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (Ø.W.); (R.L.)
| | - Kimberley Joanne Hatfield
- Department of Transfusion Medicine and Immunology, Haukeland University Hospital, 5021 Bergen, Norway;
| | - Øystein Wendelbo
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (Ø.W.); (R.L.)
| | - Roald Lindås
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (Ø.W.); (R.L.)
| | - Philippe Lassalle
- Inserm, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, U1019-UMR9017, University of Lille, 59000 Lille, France;
- Center for Infection and Immunity, le Centre Nationale de la Recherche Scientifique, Univeristy of Lille, 59000 Lille, France
- Centre d’Infection et d’Immunité de Lille, Equipe Immunité Pulmonaire, University of Lille, 59000 Lille, France
| | - Øystein Bruserud
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway;
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (Ø.W.); (R.L.)
- Correspondence:
| |
Collapse
|
33
|
Li M, Lan F, Li C, Li N, Chen X, Zhong Y, Yang Y, Shao Y, Kong Y, Li X, Wu D, Zhang J, Chen W, Li Z, Zhu X. Expression and Regulation Network of HDAC3 in Acute Myeloid Leukemia and the Implication for Targeted Therapy Based on Multidataset Data Mining. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:4703524. [PMID: 35371279 PMCID: PMC8966751 DOI: 10.1155/2022/4703524] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 02/15/2022] [Indexed: 02/07/2023]
Abstract
Background Histone deacetylase 3 (HDAC3) plays an important role in the development and progression of a variety of cancers, but its regulatory mechanism in acute myeloid leukemia (LAML) is not entirely understood. Methods We analyzed the expression of HDAC3 in normal and cancerous tissues using Oncomine, UALCAN, and GEO databases. Changes of the HDAC3 gene were analyzed by cBioPortal. The genes coexpressed with HDAC3 were analyzed by WebGestalt, and the predicted signaling pathways in KEGG were discussed. Results We discovered that the expression of HDAC3 was elevated in some types of acute myeloid leukemia. The HDAC3 gene has a strong positive correlation with SLC25A5, NDUFA2, Cox4I1, and EIF3K, which regulate cell growth and development. HDAC3 transcription is higher in patients with FLT3 mutation than in healthy people. HDAC3 can be directly involved in regulating the thyroid hormone signaling pathway. MEF2D is directly involved in the cGMP-PKG signaling pathway, and the HDAC3 gene has a strong synergistic relationship with MEF2D. HDAC3 is indirectly involved in the cGMP-PKG signaling pathway, thereby indirectly regulating the expression levels of p53 and p21 genes in patients with LAML. Genomics of Drug Sensitivity in Cancer (GDSC) database analysis revealed that the application of the HDAC3 inhibitor can inhibit the proliferation of leukemia cells. Conclusions Therefore, our data suggest that HDAC3 may be a possible therapeutic target for acute myeloid leukemia.
Collapse
Affiliation(s)
- Minhua Li
- School of Laboratory Medicine, Hangzhou Medical College, Hangzhou, China
- Zhu's Group, Guangdong Medical University, Zhanjiang, China
| | - Feifei Lan
- Medical Genetics Center, Guangdong Women and Children Hospital, Guangzhou, China
| | - Chen Li
- Department of Biology, Chemistry, Pharmacy, Free University of Berlin, 14195 Berlin, Germany
| | - Ning Li
- Zhu's Group, Guangdong Medical University, Zhanjiang, China
| | - Xiaojie Chen
- Zhu's Group, Guangdong Medical University, Zhanjiang, China
| | - Yueyuan Zhong
- Zhu's Group, Guangdong Medical University, Zhanjiang, China
| | - Yue Yang
- Zhu's Group, Guangdong Medical University, Zhanjiang, China
| | - Yingqi Shao
- Zhu's Group, Guangdong Medical University, Zhanjiang, China
| | - Yi Kong
- Zhu's Group, Guangdong Medical University, Zhanjiang, China
| | - Xinming Li
- Zhu's Group, Guangdong Medical University, Zhanjiang, China
| | - Danny Wu
- Zhu's Group, Guangdong Medical University, Zhanjiang, China
| | - Jingyu Zhang
- Zhu's Group, Guangdong Medical University, Zhanjiang, China
| | - Wenqing Chen
- Zhu's Group, Guangdong Medical University, Zhanjiang, China
| | - Zesong Li
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Shenzhen, China
| | - Xiao Zhu
- School of Laboratory Medicine, Hangzhou Medical College, Hangzhou, China
- Zhu's Group, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
34
|
Colombo M, Norfo R, Bianchi G, Roccaro AM. Editorial: The Bone Marrow Niche in Normal and Malignant Haematopoiesis. Front Cell Dev Biol 2022; 10:870114. [PMID: 35295852 PMCID: PMC8918610 DOI: 10.3389/fcell.2022.870114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 02/11/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Michela Colombo
- Haematopoietic Stem Cell Biology Laboratory, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
- *Correspondence: Michela Colombo,
| | - Ruggiero Norfo
- Haematopoietic Stem Cell Biology Laboratory, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Giada Bianchi
- Dana–Farber Cancer Institute, Boston, MA, United States
| | - Aldo M. Roccaro
- Clinical Research Development and Phase I Unit, ASST Spedali Civili di Brescia, Brescia, Italy
| |
Collapse
|