1
|
Chen ZG, Xie YT, Yang C, Xiao T, Chen SY, Wu JH, Guo QN, Gao L. M2 macrophages secrete CCL20 to regulate iron metabolism and promote daunorubicin resistance in AML cells. Life Sci 2025; 361:123297. [PMID: 39645162 DOI: 10.1016/j.lfs.2024.123297] [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: 08/20/2024] [Revised: 11/22/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024]
Abstract
Chemotherapy resistance is a significant clinical challenge in the treatment of leukemia. M2 macrophages have been identified as key contributors to the development of chemotherapy resistance in cancer, yet the precise mechanisms by which macrophages regulate this resistance remain elusive. Our study has identified CCL20 as a pivotal factor in the promotion of chemoresistance in AML cells by M2 macrophages. The chemotherapeutic agent daunorubicin induces a marked increase in ROS and lipid peroxidation levels within AML cells. This is accompanied by the inhibition of the SLC7A11/GCL/GPX4 signaling axis, elevated levels of intracellular free iron, disrupted iron metabolism, and consequent mitochondrial damage, ultimately leading to ferroptosis. Notably, CCL20 enhances the ability of AML cells to maintain iron homeostasis by upregulating SLC7A11 protein activity, mitigating mitochondrial damage, and inhibiting ferroptosis, thereby contributing to chemotherapy resistance. Furthermore, in vivo experiments demonstrated that blocking CCL20 effectively restores the sensitivity of AML cells to daunorubicin chemotherapy. Collectively, these findings underscore the complex interplay between M2 macrophages, CCL20 signaling, and chemotherapy resistance in AML, highlighting potential therapeutic avenues for intervention.
Collapse
Affiliation(s)
- Zhi-Gang Chen
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, China 400037
| | - Yu-Tong Xie
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, China 400037
| | - Chao Yang
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, China 400037
| | - Tong Xiao
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, China 400037
| | - Si-Yu Chen
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, China 400037
| | - Jun-Hong Wu
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, China 400037
| | - Qiao-Nan Guo
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, China 400037
| | - Lei Gao
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, China 400037.
| |
Collapse
|
2
|
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
|
3
|
Travaglini S, Gurnari C, Ottone T, Voso MT. Advances in the pathogenesis of FLT3 -mutated acute myeloid leukemia and targeted treatments. Curr Opin Oncol 2024; 36:569-576. [PMID: 39246183 PMCID: PMC11460763 DOI: 10.1097/cco.0000000000001094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2024]
Abstract
PURPOSE OF REVIEW FLT3 mutations are among the most common myeloid drivers identified in adult acute myeloid leukemia (AML). Their identification is crucial for the precise risk assessment because of the strong prognostic significance of the most recurrent type of FLT3 alterations, namely internal tandem duplications (ITDs). Recent advances in the pathogenesis and biology of FLT3 -mutated AML have opened an opportunity for development and application of selective inhibition of FLT3 pathway. RECENT FINDINGS In the last decade, at least three targeted treatments have been approved by regulatory agencies and several others are currently under investigations. Here, we review the latest advance in the role of FLT3 mutations in AML, providing an outline of the available therapeutic strategies, their mechanisms of actions and of resistance, as well as routes for potential improvement. SUMMARY The availability of FLT3 inhibitors has improved outcomes in AML harboring such mutations, currently also reflected in disease stratification and recommendations. Newer inhibitors are under investigations, and combinations with chemotherapy or other targeted treatments are being explored to further improve disease outcomes.
Collapse
Affiliation(s)
- Serena Travaglini
- Department of Biomedicine and Prevention, University of Tor Vergata
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Carmelo Gurnari
- Department of Biomedicine and Prevention, University of Tor Vergata
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Tiziana Ottone
- Department of Biomedicine and Prevention, University of Tor Vergata
| | | |
Collapse
|
4
|
Zhou Y, Huang G, Cai X, Liu Y, Qian B, Li D. Global, regional, and national burden of acute myeloid leukemia, 1990-2021: a systematic analysis for the global burden of disease study 2021. Biomark Res 2024; 12:101. [PMID: 39256810 PMCID: PMC11389310 DOI: 10.1186/s40364-024-00649-y] [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: 08/24/2024] [Accepted: 08/31/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML), as the most common subtype of leukemia in adults, is characterised by rapid progression and poor prognosis. In the context of the rapid development of medical technology and the complexity of social factors, a detailed report describing the latest epidemiological patterns of AML is important for decision makers to allocate healthcare resources effectively. METHODS Our research utilized the latest data sourced from the Global Burden of Disease (GBD) 2021. To delineate the burden of AML, we comprehensively described the incidence, deaths, disability-adjusted life years (DALYs), and the associated age-standardized rates per 100,000 persons (ASR) spanning from 1990 to 2021 stratifies according to age, sex, socio-demographic index (SDI), and nationality. Additionally, we extracted and analyzed data about the risk factors that contribute to AML-related deaths and DALYs. RESULTS According to our study, the incidence of AML has continued to rise globally from 79,372 in 1990 to 144,645 in 2021 and AML affected the male and the elderly populations disproportionately. Furthermore, there was a significant positive correlation between the burden of AML and the SDI value. Developed nations generally exhibited higher age-standardized incidence rate, age-standardized death rate, and age-standardized disability-adjusted life year rate than the developing nations. We also analyzed the prevalence of smoking, high body mass index, and occupational benzene and formaldehyde exposure in the AML population in different SDI regions. Moreover, smoking and high body mass index were more prevalent in developed countries, whereas occupational exposure to these chemicals was the predominant risk factor in developing countries. CONCLUSION The global burden of AML has increased over the past 32 years, with rising morbidity and mortality. The incidence of AML is differentially distributed across different SDI countries or regions. AML incidence is higher in the elderly and in men. The proportions of smoking, high body mass index, and occupational exposure to benzene and formaldehyde varied by region. The findings highlight the need for region-specific prevention and call for future research on preventive strategies and new treatments to lower AML incidence and improve patient outcomes.
Collapse
Affiliation(s)
- Yeming Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guiqin Huang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoya Cai
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Liu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bingxin Qian
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dengju Li
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
5
|
Spertini C, Bénéchet AP, Birch F, Bellotti A, Román-Trufero M, Arber C, Auner HW, Mitchell RA, Spertini O, Smirnova T. Macrophage migration inhibitory factor blockade reprograms macrophages and disrupts prosurvival signaling in acute myeloid leukemia. Cell Death Discov 2024; 10:157. [PMID: 38548753 PMCID: PMC10978870 DOI: 10.1038/s41420-024-01924-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 03/14/2024] [Accepted: 03/19/2024] [Indexed: 04/01/2024] Open
Abstract
The malignant microenvironment plays a major role in the development of resistance to therapies and the occurrence of relapses in acute myeloid leukemia (AML). We previously showed that interactions of AML blasts with bone marrow macrophages (MΦ) shift their polarization towards a protumoral (M2-like) phenotype, promoting drug resistance; we demonstrated that inhibiting the colony-stimulating factor-1 receptor (CSF1R) repolarizes MΦ towards an antitumoral (M1-like) phenotype and that other factors may be involved. We investigated here macrophage migration inhibitory factor (MIF) as a target in AML blast survival and protumoral interactions with MΦ. We show that pharmacologically inhibiting MIF secreted by AML blasts results in their apoptosis. However, this effect is abrogated when blasts are co-cultured in close contact with M2-like MΦ. We next demonstrate that pharmacological inhibition of MIF secreted by MΦ, in the presence of granulocyte macrophage-colony stimulating factor (GM-CSF), efficiently reprograms MΦ to an M1-like phenotype that triggers apoptosis of interacting blasts. Furthermore, contact with reprogrammed MΦ relieves blast resistance to venetoclax and midostaurin acquired in contact with CD163+ protumoral MΦ. Using intravital imaging in mice, we also show that treatment with MIF inhibitor 4-IPP and GM-CSF profoundly affects the tumor microenvironment in vivo: it strikingly inhibits tumor vasculature, reduces protumoral MΦ, and slows down leukemia progression. Thus, our data demonstrate that MIF plays a crucial role in AML MΦ M2-like protumoral phenotype that can be reversed by inhibiting its activity and suggest the therapeutic targeting of MIF as an avenue towards improved AML treatment outcomes.
Collapse
Affiliation(s)
- Caroline Spertini
- Service and Central Laboratory of Hematology, Lausanne University Hospital (CHUV), 1011, Lausanne, Switzerland
| | - Alexandre P Bénéchet
- In Vivo Imaging Facility (IVIF), Department of Research and Training, Lausanne University Hospital and University of Lausanne, Lausanne, 1011, Switzerland
| | - Flora Birch
- Department of oncology UNIL-CHUV, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), 1011, Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, 1015, Lausanne, Switzerland
| | - Axel Bellotti
- Service and Central Laboratory of Hematology, Lausanne University Hospital (CHUV), 1011, Lausanne, Switzerland
| | - Mónica Román-Trufero
- Service and Central Laboratory of Hematology, Lausanne University Hospital (CHUV), 1011, Lausanne, Switzerland
| | - Caroline Arber
- Service and Central Laboratory of Hematology, Lausanne University Hospital (CHUV), 1011, Lausanne, Switzerland
- Department of oncology UNIL-CHUV, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), 1011, Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, 1015, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, 1011, Lausanne, Switzerland
- Service of Immuno-oncology, Lausanne University Hospital (CHUV), 1011, Lausanne, Switzerland
| | - Holger W Auner
- Service and Central Laboratory of Hematology, Lausanne University Hospital (CHUV), 1011, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, 1011, Lausanne, Switzerland
| | - Robert A Mitchell
- Department of Surgery, Division of Immunotherapy, University of Louisville, Louisville, KY, 40202, USA
| | - Olivier Spertini
- Faculty of Biology and Medicine, University of Lausanne, 1011, Lausanne, Switzerland
| | - Tatiana Smirnova
- Service and Central Laboratory of Hematology, Lausanne University Hospital (CHUV), 1011, Lausanne, Switzerland.
| |
Collapse
|
6
|
Kolesnikova MA, Sen’kova AV, Pospelova TI, Zenkova MA. Effective Prognostic Model for Therapy Response Prediction in Acute Myeloid Leukemia Patients. J Pers Med 2023; 13:1234. [PMID: 37623484 PMCID: PMC10455213 DOI: 10.3390/jpm13081234] [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: 06/30/2023] [Revised: 07/29/2023] [Accepted: 08/05/2023] [Indexed: 08/26/2023] Open
Abstract
Acute myeloid leukemia (AML) is a hematopoietic disorder characterized by the malignant transformation of bone marrow-derived myeloid progenitor cells with extremely short survival. To select the optimal treatment options and predict the response to therapy, the stratification of AML patients into risk groups based on genetic factors along with clinical characteristics is carried out. Despite this thorough approach, the therapy response and disease outcome for a particular patient with AML depends on several patient- and tumor-associated factors. Among these, tumor cell resistance to chemotherapeutic agents represents one of the main obstacles for improving survival outcomes in AML patients. In our study, a new prognostic scale for the risk stratification of AML patients based on the detection of the sensitivity or resistance of tumor cells to chemotherapeutic drugs in vitro as well as MDR1 mRNA/P-glycoprotein expression, tumor origin (primary or secondary), cytogenetic abnormalities, and aberrant immunophenotype was developed. This study included 53 patients diagnosed with AML. Patients who received intensive or non-intensive induction therapy were analyzed separately. Using correlation, ROC, and Cox regression analyses, we show that the risk stratification of AML patients in accordance with the developed prognostic scale correlates well with the response to therapy and represents an independent predictive factor for the overall survival of patients with newly diagnosed AML.
Collapse
Affiliation(s)
| | - Aleksandra V. Sen’kova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk 630090, Russia;
| | - Tatiana I. Pospelova
- Department of Therapy, Hematology and Transfusiology, Novosibirsk State Medical University, Novosibirsk 630091, Russia;
| | - Marina A. Zenkova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk 630090, Russia;
| |
Collapse
|
7
|
Franza M, Albanesi J, Mancini B, Pennisi R, Leone S, Acconcia F, Bianchi F, di Masi A. The clinically relevant CHK1 inhibitor MK-8776 induces the degradation of the oncogenic protein PML-RARα and overcomes ATRA resistance in acute promyelocytic leukemia cells. Biochem Pharmacol 2023:115675. [PMID: 37406967 DOI: 10.1016/j.bcp.2023.115675] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 06/27/2023] [Accepted: 06/27/2023] [Indexed: 07/07/2023]
Abstract
Acute promyelocytic leukemia (APL) is a hematological disease characterized by the expression of the oncogenic fusion protein PML-RARα. The current treatment approach for APL involves differentiation therapy using all-trans retinoic acid (ATRA) and arsenic trioxide (ATO). However, the development of resistance to therapy, occurrence of differentiation syndrome, and relapses necessitate the exploration of new treatment options that induce differentiation of leukemic blasts with low toxicity. In this study, we investigated the cellular and molecular effects of MK-8776, a specific inhibitor of CHK1, in ATRA-resistant APL cells. Treatment of APL cells with MK-8776 resulted in a decrease in PML-RARα levels, increased expression of CD11b, and increased granulocytic activity consistent with differentiation. Interestingly, we showed that the MK-8776-induced differentiating effect resulted synergic with ATO. We found that the reduction of PML-RARα by MK-8776 was dependent on both proteasome and caspases. Specifically, both caspase-1 and caspase-3 were activated by CHK1 inhibition, with caspase-3 acting upstream of caspase-1. Activation of caspase-3 was necessary to activate caspase-1 and promote PML-RARα degradation. Transcriptomic analysis revealed significant modulation of pathways and upstream regulators involved in the inflammatory response and cell cycle control upon MK-8776 treatment. Overall, the ability of MK-8776 to induce PML-RARα degradation and stimulate differentiation of immature APL cancer cells into more mature forms recapitulates the concept of differentiation therapy. Considering the in vivo tolerability of MK-8776, it will be relevant to evaluate its potential clinical benefit in APL patients resistant to standard ATRA/ATO therapy, as well as in patients with other forms of acute leukemias.
Collapse
Affiliation(s)
- Maria Franza
- Department of Sciences, Section of Biomedical Sciences and Technologies, Roma Tre University, Roma, Italy
| | - Jacopo Albanesi
- Department of Sciences, Section of Biomedical Sciences and Technologies, Roma Tre University, Roma, Italy
| | - Benedetta Mancini
- Department of Sciences, Section of Biomedical Sciences and Technologies, Roma Tre University, Roma, Italy
| | - Rosa Pennisi
- Department of Oncology, University of Torino Medical School, Torino, Italy; Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Torino, Italy
| | - Stefano Leone
- Department of Sciences, Section of Biomedical Sciences and Technologies, Roma Tre University, Roma, Italy
| | - Filippo Acconcia
- Department of Sciences, Section of Biomedical Sciences and Technologies, Roma Tre University, Roma, Italy
| | - Fabrizio Bianchi
- Unit of Cancer Biomarkers, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Alessandra di Masi
- Department of Sciences, Section of Biomedical Sciences and Technologies, Roma Tre University, Roma, Italy.
| |
Collapse
|
8
|
Pagliuca S, Gurnari C, Zhang K, Kewan T, Bahaj W, Mori M, Nautiyal I, Rubio MT, Ferraro F, Maciejewski JP, Wang L, Visconte V. Comprehensive Transcriptomic Analysis of VISTA in Acute Myeloid Leukemia: Insights into Its Prognostic Value. Int J Mol Sci 2022; 23:ijms232314885. [PMID: 36499220 PMCID: PMC9735915 DOI: 10.3390/ijms232314885] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/22/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
The V-domain Ig suppressor of T-cell activation (VISTA) has been recognized as a critical negative regulator of antitumor immune response and is gaining growing interest as a potential pharmacological target in immunotherapy. This molecule is highly expressed in hematopoietic stem cells and myeloid compartment, and it has been found upmodulated in acute myeloid leukemia (AML). However, VISTA-associated immune features are relatively unexplored in myeloid malignancies. Herein, we aimed to explore whether this immune checkpoint regulator could play a role in the generation of an immune escape environment in AML patients. We characterized VISTA mRNA expression levels in leukemia cell lines and in large publicly available cohorts of specimens from bone marrow of healthy individuals and AML patients at diagnosis by deploying bulk and single-cell RNA sequencing. We also defined the correlations with leukemia-associated burden using results of whole-exome sequencing of AML samples at disease onset. We showed that VISTA expression linearly increased across the myeloid differentiation tree in normal hematopoiesis. Accordingly, its transcript was highly enriched in AML cell lines as well as in AML patients at diagnosis presenting with myelomonocytic and monocytic differentiation. A strong correlation was seen with NPM1 mutations regardless of the presence of FLT3 lesions. Furthermore, VISTA expression levels at baseline correlated with disease recurrence in patients with normal karyotype and NPM1 mutations, a subgroup traditionally considered as favorable according to current diagnostic schemes. Indeed, when compared to patients with long-term remission (>5 years after standard chemotherapy regimens), cases relapsing within 2 years from diagnosis had increased VISTA expression in both leukemia and T cells. Our results suggest a rationale for developing VISTA-targeted therapeutic strategies to treat molecularly defined subgroups of AML patients to prevent disease recurrence and treatment resistance.
Collapse
Affiliation(s)
- Simona Pagliuca
- Translational Hematology and Oncology Research Department of Cleveland Clinic, Cleveland, OH 44106, USA
- Service d’hématologie, Hôpital Brabois, CHRU Nancy and CNRS UMR 7365 IMoPa, Biopôle de l’Université de Lorraine, 54500 Vandoeuvre les Nancy, France
| | - Carmelo Gurnari
- Translational Hematology and Oncology Research Department of Cleveland Clinic, Cleveland, OH 44106, USA
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Keman Zhang
- Translational Hematology and Oncology Research Department of Cleveland Clinic, Cleveland, OH 44106, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
| | - Tariq Kewan
- Translational Hematology and Oncology Research Department of Cleveland Clinic, Cleveland, OH 44106, USA
| | - Waled Bahaj
- Translational Hematology and Oncology Research Department of Cleveland Clinic, Cleveland, OH 44106, USA
| | - Minako Mori
- Translational Hematology and Oncology Research Department of Cleveland Clinic, Cleveland, OH 44106, USA
| | - Ishani Nautiyal
- Translational Hematology and Oncology Research Department of Cleveland Clinic, Cleveland, OH 44106, USA
| | - Marie Thérèse Rubio
- Service d’hématologie, Hôpital Brabois, CHRU Nancy and CNRS UMR 7365 IMoPa, Biopôle de l’Université de Lorraine, 54500 Vandoeuvre les Nancy, France
| | - Francesca Ferraro
- Division of Oncology, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Jaroslaw P. Maciejewski
- Translational Hematology and Oncology Research Department of Cleveland Clinic, Cleveland, OH 44106, USA
| | - Li Wang
- Translational Hematology and Oncology Research Department of Cleveland Clinic, Cleveland, OH 44106, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
| | - Valeria Visconte
- Translational Hematology and Oncology Research Department of Cleveland Clinic, Cleveland, OH 44106, USA
- Correspondence:
| |
Collapse
|
9
|
A Focus on Intermediate-Risk Acute Myeloid Leukemia: Sub-Classification Updates and Therapeutic Challenges. Cancers (Basel) 2022; 14:cancers14174166. [PMID: 36077703 PMCID: PMC9454629 DOI: 10.3390/cancers14174166] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/16/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022] Open
Abstract
Acute myeloid leukemia (AML) represents a heterogeneous group of hematopoietic neoplasms deriving from the abnormal proliferation of myeloid progenitors in the bone marrow. Patients with AML may have highly variable outcomes, which are generally dictated by individual clinical and genomic characteristics. As such, the European LeukemiaNet 2017 and 2022 guidelines categorize newly diagnosed AML into favorable-, intermediate-, and adverse-risk groups, based on their molecular and cytogenetic profiles. Nevertheless, the intermediate-risk category remains poorly defined, as many patients fall into this group as a result of their exclusion from the other two. Moreover, further genomic data with potential prognostic and therapeutic influences continue to emerge, though they are yet to be integrated into the diagnostic and prognostic models of AML. This review highlights the latest therapeutic advances and challenges that warrant refining the prognostic classification of intermediate-risk AML.
Collapse
|
10
|
Fiedler W, Montesinos P, Schliemann C, Middeke J, Vasu S, Scholz CW, Esteve J, Mondal S, Rüter B, Burkard U, Osswald A, Blum W. An open-label, phase I/II trial to determine the maximum tolerated dose and investigate safety, pharmacokinetics and efficacy of BI 836858, an unconjugated anti-CD33 monoclonal antibody, in combination with decitabine in patients with acute myeloid leukemia. Haematologica 2022; 107:2977-2982. [PMID: 36005556 PMCID: PMC9713566 DOI: 10.3324/haematol.2022.281128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Indexed: 12/14/2022] Open
Affiliation(s)
- Walter Fiedler
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Pau Montesinos
- Hospital Universitari i Politècnic La Fe, Valencia, Spain,CIBERONC, Instituto Carlos III, Madrid, Spain
| | - Christoph Schliemann
- Department of Medicine A, Hematology and Oncology, University Hospital Muenster, Muenster, Germany
| | | | - Sumithira Vasu
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Christian W. Scholz
- Department of Hematology and Oncology, Vivantes Klinikum Am Urban, Berlin, Germany
| | - Jordi Esteve
- Hematology Department, Hospital Clínic of Barcelona, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Shoubhik Mondal
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA
| | - Björn Rüter
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach/Riss, Germany and
| | - Ute Burkard
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach/Riss, Germany and
| | - Annika Osswald
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach/Riss, Germany and
| | - William Blum
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA,W. BLUM -
| |
Collapse
|
11
|
Morell A, Budagaga Y, Vagiannis D, Zhang Y, Laštovičková L, Novotná E, Haddad A, Haddad M, Portillo R, Hofman J, Wsól V. Isocitrate dehydrogenase 2 inhibitor enasidenib synergizes daunorubicin cytotoxicity by targeting aldo-keto reductase 1C3 and ATP-binding cassette transporters. Arch Toxicol 2022; 96:3265-3277. [PMID: 35972551 DOI: 10.1007/s00204-022-03359-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 08/03/2022] [Indexed: 12/01/2022]
Abstract
Targeting mutations that trigger acute myeloid leukaemia (AML) has emerged as a refined therapeutic approach in recent years. Enasidenib (Idhifa) is the first selective inhibitor of mutated forms of isocitrate dehydrogenase 2 (IDH2) approved against relapsed/refractory AML. In addition to its use as monotherapy, a combination trial of enasidenib with standard intensive induction therapy (daunorubicin + cytarabine) is being evaluated. This study aimed to decipher enasidenib off-target molecular mechanisms involved in anthracycline resistance, such as reduction by carbonyl reducing enzymes (CREs) and drug efflux by ATP-binding cassette (ABC) transporters. We analysed the effect of enasidenib on daunorubicin (Daun) reduction by several recombinant CREs and different human cell lines expressing aldo-keto reductase 1C3 (AKR1C3) exogenously (HCT116) or endogenously (A549 and KG1a). Additionally, A431 cell models overexpressing ABCB1, ABCG2, or ABCC1 were employed to evaluate enasidenib modulation of Daun efflux. Furthermore, the potential synergism of enasidenib over Daun cytotoxicity was quantified amongst all the cell models. Enasidenib selectively inhibited AKR1C3-mediated inactivation of Daun in vitro and in cell lines expressing AKR1C3, as well as its extrusion by ABCB1, ABCG2, and ABCC1 transporters, thus synergizing Daun cytotoxicity to overcome resistance. This work provides in vitro evidence on enasidenib-mediated targeting of the anthracycline resistance actors AKR1C3 and ABC transporters under clinically achievable concentrations. Our findings may encourage its combination with intensive chemotherapy and even suggest that the effectiveness of enasidenib as monotherapy against AML could lie beyond the targeting of mIDH2.
Collapse
Affiliation(s)
- Anselm Morell
- Department of Biochemical Sciences, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Youssif Budagaga
- Department of Pharmacology, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Dimitrios Vagiannis
- Department of Pharmacology, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Yu Zhang
- Department of Pharmacology, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Lenka Laštovičková
- Department of Biochemical Sciences, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Eva Novotná
- Department of Biochemical Sciences, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Andrew Haddad
- Department of Biochemical Sciences, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Melodie Haddad
- Department of Biochemical Sciences, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Ramon Portillo
- Department of Pharmacology, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Jakub Hofman
- Department of Pharmacology, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Vladimír Wsól
- Department of Biochemical Sciences, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic.
| |
Collapse
|
12
|
Ancos-Pintado R, Bragado-García I, Morales ML, García-Vicente R, Arroyo-Barea A, Rodríguez-García A, Martínez-López J, Linares M, Hernández-Sánchez M. High-Throughput CRISPR Screening in Hematological Neoplasms. Cancers (Basel) 2022; 14:3612. [PMID: 35892871 PMCID: PMC9329962 DOI: 10.3390/cancers14153612] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/16/2022] [Accepted: 07/20/2022] [Indexed: 02/01/2023] Open
Abstract
CRISPR is becoming an indispensable tool in biological research, revolutionizing diverse fields of medical research and biotechnology. In the last few years, several CRISPR-based genome-targeting tools have been translated for the study of hematological neoplasms. However, there is a lack of reviews focused on the wide uses of this technology in hematology. Therefore, in this review, we summarize the main CRISPR-based approaches of high throughput screenings applied to this field. Here we explain several libraries and algorithms for analysis of CRISPR screens used in hematology, accompanied by the most relevant databases. Moreover, we focus on (1) the identification of novel modulator genes of drug resistance and efficacy, which could anticipate relapses in patients and (2) new therapeutic targets and synthetic lethal interactions. We also discuss the approaches to uncover novel biomarkers of malignant transformations and immune evasion mechanisms. We explain the current literature in the most common lymphoid and myeloid neoplasms using this tool. Then, we conclude with future directions, highlighting the importance of further gene candidate validation and the integration and harmonization of the data from CRISPR screening approaches.
Collapse
Affiliation(s)
- Raquel Ancos-Pintado
- Department of Translational Hematology, Instituto de Investigación Hospital 12 de Octubre (imas12), Hematological Malignancies Clinical Research Unit H12O-CNIO, CIBERONC, ES 28041 Madrid, Spain; (R.A.-P.); (M.L.M.); (R.G.-V.); (A.R.-G.); (J.M.-L.); (M.L.)
- Department of Biochemistry and Molecular Biology, Pharmacy School, Universidad Complutense de Madrid, ES 28040 Madrid, Spain; (I.B.-G.); (A.A.-B.)
| | - Irene Bragado-García
- Department of Biochemistry and Molecular Biology, Pharmacy School, Universidad Complutense de Madrid, ES 28040 Madrid, Spain; (I.B.-G.); (A.A.-B.)
| | - María Luz Morales
- Department of Translational Hematology, Instituto de Investigación Hospital 12 de Octubre (imas12), Hematological Malignancies Clinical Research Unit H12O-CNIO, CIBERONC, ES 28041 Madrid, Spain; (R.A.-P.); (M.L.M.); (R.G.-V.); (A.R.-G.); (J.M.-L.); (M.L.)
| | - Roberto García-Vicente
- Department of Translational Hematology, Instituto de Investigación Hospital 12 de Octubre (imas12), Hematological Malignancies Clinical Research Unit H12O-CNIO, CIBERONC, ES 28041 Madrid, Spain; (R.A.-P.); (M.L.M.); (R.G.-V.); (A.R.-G.); (J.M.-L.); (M.L.)
| | - Andrés Arroyo-Barea
- Department of Biochemistry and Molecular Biology, Pharmacy School, Universidad Complutense de Madrid, ES 28040 Madrid, Spain; (I.B.-G.); (A.A.-B.)
| | - Alba Rodríguez-García
- Department of Translational Hematology, Instituto de Investigación Hospital 12 de Octubre (imas12), Hematological Malignancies Clinical Research Unit H12O-CNIO, CIBERONC, ES 28041 Madrid, Spain; (R.A.-P.); (M.L.M.); (R.G.-V.); (A.R.-G.); (J.M.-L.); (M.L.)
| | - Joaquín Martínez-López
- Department of Translational Hematology, Instituto de Investigación Hospital 12 de Octubre (imas12), Hematological Malignancies Clinical Research Unit H12O-CNIO, CIBERONC, ES 28041 Madrid, Spain; (R.A.-P.); (M.L.M.); (R.G.-V.); (A.R.-G.); (J.M.-L.); (M.L.)
- Department of Medicine, Medicine School, Universidad Complutense de Madrid, ES 28040 Madrid, Spain
| | - María Linares
- Department of Translational Hematology, Instituto de Investigación Hospital 12 de Octubre (imas12), Hematological Malignancies Clinical Research Unit H12O-CNIO, CIBERONC, ES 28041 Madrid, Spain; (R.A.-P.); (M.L.M.); (R.G.-V.); (A.R.-G.); (J.M.-L.); (M.L.)
- Department of Biochemistry and Molecular Biology, Pharmacy School, Universidad Complutense de Madrid, ES 28040 Madrid, Spain; (I.B.-G.); (A.A.-B.)
| | - María Hernández-Sánchez
- Department of Biochemistry and Molecular Biology, Pharmacy School, Universidad Complutense de Madrid, ES 28040 Madrid, Spain; (I.B.-G.); (A.A.-B.)
| |
Collapse
|
13
|
Immunoproteasome Activity in Chronic Lymphocytic Leukemia as a Target of the Immunoproteasome-Selective Inhibitors. Cells 2022; 11:cells11050838. [PMID: 35269460 PMCID: PMC8909520 DOI: 10.3390/cells11050838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/21/2022] [Accepted: 02/22/2022] [Indexed: 12/21/2022] Open
Abstract
Targeting proteasome with proteasome inhibitors (PIs) is an approved treatment strategy in multiple myeloma that has also been explored pre-clinically and clinically in other hematological malignancies. The approved PIs target both the constitutive and the immunoproteasome, the latter being present predominantly in cells of lymphoid origin. Therapeutic targeting of the immunoproteasome in cells with sole immunoproteasome activity may be selectively cytotoxic in malignant cells, while sparing the non-lymphoid tissues from the on-target PIs toxicity. Using activity-based probes to assess the proteasome activity profile and correlating it with the cytotoxicity assays, we identified B-cell chronic lymphocytic leukemia (B-CLL) to express predominantly immunoproteasome activity, which is associated with high sensitivity to approved proteasome inhibitors and, more importantly, to the immunoproteasome selective inhibitors LU005i and LU035i, targeting all immunoproteasome active subunits or only the immunoproteasome β5i, respectively. At the same time, LU102, a proteasome β2 inhibitor, sensitized B-CLL or immunoproteasome inhibitor-inherently resistant primary cells of acute myeloid leukemia, B-cell acute lymphoblastic leukemia, multiple myeloma and plasma cell leukemia to low doses of LU035i. The immunoproteasome thus represents a novel therapeutic target, which warrants further testing with clinical stage immunoproteasome inhibitors in monotherapy or in combinations.
Collapse
|
14
|
Jordheim LP. Toward Personalization of Cytarabine Dosing in Acute Myeloid Leukemia. J Clin Oncol 2022; 40:784-786. [PMID: 35025625 DOI: 10.1200/jco.21.02723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Lars Petter Jordheim
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| |
Collapse
|
15
|
CSF1R Inhibition Combined with GM-CSF Reprograms Macrophages and Disrupts Protumoral Interplays with AML Cells. Cancers (Basel) 2021; 13:cancers13215289. [PMID: 34771453 PMCID: PMC8582394 DOI: 10.3390/cancers13215289] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/04/2021] [Accepted: 10/15/2021] [Indexed: 12/20/2022] Open
Abstract
Relapse is a major issue in acute myeloid leukemia (AML) and while the contribution of gene mutations in developing drug resistance is well established, little is known on the role of macrophages (MΦs) in an AML cell microenvironment. We examined whether myeloblasts could educate MΦs to adopt a protumoral orientation supporting myeloblast survival and resistance to therapy. Flow cytometry analyses demonstrated that M2-like CD163+ MΦs are abundantly present, at diagnosis, in the bone marrow of AML patients. We showed that myeloblasts, or their conditioned medium, polarize monocytes to M2-like CD163+ MΦs, induce the secretion of many protumoral factors, and promote myeloblast survival and proliferation as long as close intercellular contacts are maintained. Importantly, pharmacologic inhibition of the CSF1 receptor (CSF1R), in the presence of GM-CSF, reprogrammed MΦ polarization to an M1-like orientation, induced the secretion of soluble factors with antitumoral activities, reduced protumoral agonists, and promoted the apoptosis of myeloblasts interacting with MΦs. Furthermore, myeloblasts, which became resistant to venetoclax or midostaurin during their interplay with protumoral CD163+ MΦs, regained sensitivity to these targeted therapies following CSF1R inhibition in the presence of GM-CSF. These data reveal a crucial role of CD163+ MΦ interactions with myeloblasts that promote myeloblast survival and identify CSF1R inhibition as a novel target for AML therapy.
Collapse
|
16
|
Karakitsou E, Foguet C, Contreras Mostazo MG, Kurrle N, Schnütgen F, Michaelis M, Cinatl J, Marin S, Cascante M. Genome-scale integration of transcriptome and metabolome unveils squalene synthase and dihydrofolate reductase as targets against AML cells resistant to chemotherapy. Comput Struct Biotechnol J 2021; 19:4059-4066. [PMID: 34377370 PMCID: PMC8326745 DOI: 10.1016/j.csbj.2021.06.049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/29/2021] [Accepted: 06/29/2021] [Indexed: 01/06/2023] Open
Abstract
The development of resistance to chemotherapeutic agents, such as Doxorubicin (DOX) and cytarabine (AraC), is one of the greatest challenges to the successful treatment of Acute Myeloid Leukemia (AML). Such acquisition is often underlined by a metabolic reprogramming that can provide a therapeutic opportunity, as it can lead to the emergence of vulnerabilities and dependencies to be exploited as targets against the resistant cells. In this regard, genome-scale metabolic models (GSMMs) have emerged as powerful tools to integrate multiple layers of data to build cancer-specific models and identify putative metabolic vulnerabilities. Here, we use genome-scale metabolic modelling to reconstruct a GSMM of the THP1 AML cell line and two derivative cell lines, one with acquired resistance to AraC and the second with acquired resistance to DOX. We also explore how, adding to the transcriptomic layer, the metabolomic layer enhances the selectivity of the resulting condition specific reconstructions. The resulting models enabled us to identify and experimentally validate that drug-resistant THP1 cells are sensitive to the FDA-approved antifolate methotrexate. Moreover, we discovered and validated that the resistant cell lines could be selectively targeted by inhibiting squalene synthase, providing a new and promising strategy to directly inhibit cholesterol synthesis in AML drug resistant cells.
Collapse
Affiliation(s)
- Effrosyni Karakitsou
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Spain
- Institute of Biomedicine of University of Barcelona, 08028 Barcelona, Spain
- CIBER of Hepatic and Digestive Diseases (CIBEREHD), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| | - Carles Foguet
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Spain
- Institute of Biomedicine of University of Barcelona, 08028 Barcelona, Spain
- CIBER of Hepatic and Digestive Diseases (CIBEREHD), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
- Metabolomics Node at Spanish National Bioinformatics Institute (INB-ISCIII-ES-ELIXIR), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| | - Miriam G. Contreras Mostazo
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Spain
- Institute of Biomedicine of University of Barcelona, 08028 Barcelona, Spain
- CIBER of Hepatic and Digestive Diseases (CIBEREHD), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| | - Nina Kurrle
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe-University, 60590 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Frankfurt Cancer Institute (FCI), Goethe University, 60590 Frankfurt am Main, Germany
| | - Frank Schnütgen
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe-University, 60590 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Frankfurt Cancer Institute (FCI), Goethe University, 60590 Frankfurt am Main, Germany
| | - Martin Michaelis
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - Jindrich Cinatl
- Institut für Medizinische Virologie, Klinikum der Goethe-Universität, Frankfurt am Main, Germany
| | - Silvia Marin
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Spain
- Institute of Biomedicine of University of Barcelona, 08028 Barcelona, Spain
- CIBER of Hepatic and Digestive Diseases (CIBEREHD), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
- Metabolomics Node at Spanish National Bioinformatics Institute (INB-ISCIII-ES-ELIXIR), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| | - Marta Cascante
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Spain
- Institute of Biomedicine of University of Barcelona, 08028 Barcelona, Spain
- CIBER of Hepatic and Digestive Diseases (CIBEREHD), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
- Metabolomics Node at Spanish National Bioinformatics Institute (INB-ISCIII-ES-ELIXIR), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| |
Collapse
|
17
|
Gene Transcription as a Therapeutic Target in Leukemia. Int J Mol Sci 2021; 22:ijms22147340. [PMID: 34298959 PMCID: PMC8304797 DOI: 10.3390/ijms22147340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 12/11/2022] Open
Abstract
Blood malignancies often arise from undifferentiated hematopoietic stem cells or partially differentiated stem-like cells. A tight balance of multipotency and differentiation, cell division, and quiescence underlying normal hematopoiesis requires a special program governed by the transcriptional machinery. Acquisition of drug resistance by tumor cells also involves reprogramming of their transcriptional landscape. Limiting tumor cell plasticity by disabling reprogramming of the gene transcription is a promising strategy for improvement of treatment outcomes. Herein, we review the molecular mechanisms of action of transcription-targeted drugs in hematological malignancies (largely in leukemia) with particular respect to the results of clinical trials.
Collapse
|