1
|
Martino EA, Bruzzese A, Labanca C, Mendicino F, Lucia E, Olivito V, Stanzione G, Zimbo A, Pozzi S, Neri A, Morabito F, Vigna E, Gentile M. Investigational CXCR4 inhibitors in early phase development for the treatment of hematological malignancies. Expert Opin Investig Drugs 2024; 33:915-924. [PMID: 39096094 DOI: 10.1080/13543784.2024.2388567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 07/02/2024] [Accepted: 08/01/2024] [Indexed: 08/04/2024]
Abstract
INTRODUCTION CXCR4/CXCL12 axis regulates cell proliferation, survival, and differentiation, as well as the homing and mobilization of hematopoietic stem cells (HSCs) from bone marrow niches to the peripheral blood. Furthermore, CXCR4 and CXCL12 are key mediators of cross-talk between hematological malignancies and their microenvironments. CXCR4 overexpression drives disease progression, boosts tumor cell survival, and promotes chemoresistance, leading to poor prognosis. AREAS COVERED In light of these discoveries, scientific investigations, and clinical trials have underscored the therapeutic promise found in small-molecule antagonists like plerixafor, peptides/peptidomimetics, such as BKT140, monoclonal antibodies like PF-06747143 and ulocuplumab, as well as microRNAs. Their efficacy is evident in reducing tumor burden, inducing apoptosis and sensitizing malignant cells to conventional chemotherapies. This overview delves into the pathogenic role of the CXC4/CXCL12 axis in hematological neoplasms and examines the clinical application of key CXCR4 antagonists. EXPERT OPINION The information collectively emphasizes the potential of CXCR4 antagonists as a therapeutic strategy for hematologic malignancies, showcasing advancements in preclinical and clinical studies. As these therapeutic strategies progress through clinical trials, their potential to reshape the prognosis of hematologic malignancies will become increasingly apparent.
Collapse
Affiliation(s)
| | | | | | | | - Eugenio Lucia
- Hematology Unit, Azienda Ospedaliera Annunziata, Cosenza, Italy
| | | | - Gaia Stanzione
- Hematology Unit, Azienda Ospedaliera Annunziata, Cosenza, Italy
- Division of Hematology, Azienda Policlinico-S. Marco, University of Catania, Catania, Italy
| | - Annamaria Zimbo
- Hematology Unit, Azienda Ospedaliera Annunziata, Cosenza, Italy
- UOC Laboratorio Analisi Cliniche, Biomolecolari e Genetica, Azienda Ospedaliera Annunziata, Cosenza, Italy
| | - Stefano Pozzi
- Ematologia Azienda USL-IRCSS Reggio Emilia, Reggio Emilia, Emilia-Romagna, Italy
| | - Antonino Neri
- Scientific Directorate IRCCS of Reggio Emilia, Reggio Emilia, EmiliaRomagna, Italy
| | | | - Ernesto Vigna
- Hematology Unit, Azienda Ospedaliera Annunziata, Cosenza, Italy
| | - Massimo Gentile
- Hematology Unit, Azienda Ospedaliera Annunziata, Cosenza, Italy
- Department of Pharmacy, Health and Nutritional Science, University of Calabria, Rende, Italy
| |
Collapse
|
2
|
Yang L, Xu Q, Li J. Prognostic impact of ARHGAP43(SH3BP1) in acute myeloid leukemia. J Formos Med Assoc 2024; 123:992-1003. [PMID: 38582737 DOI: 10.1016/j.jfma.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/14/2024] [Accepted: 04/01/2024] [Indexed: 04/08/2024] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a hematological malignancy with a heterogeneous prognosis. Novel markers are required to accurately assess the prognosis and formulate treatment plans. METHODS The association of ARHGAP family genes with prognostic value in acute myeloid leukemia (AML) was assessed using public databases (CCLE, GEPIA, TCGA, and GEO). RESULTS Elevated expression of ARHGAP43 (SH3BP1) was associated with poor prognosis in patients with acute myeloid leukemia. ARHGAP43 (SH3BP1) expression was higher in the poor/adverse prognosis (P < 0.001) and TP53 mutation groups (P = 0.0093). Higher ARHGAP43 (SH3BP1) expression was found to be an independent prognostic predictor in multivariate COX regression analysis (HR = 1.317, 95% CI: 1.008-1.720, P = 0.044). Higher ARHGAP43 (SH3BP1) expression who did not receive hematopoietic stem cell transplantation (HSCT) had shorter overall survival (OS) and progression-free survival (PFS) (OS: median: 7.60 vs. 24.90 months; P = 0.006; PFS: median: 11.40 vs. 27.22 months; P = 0.0096), whereas OS and PFS of patients who received HSCT were unaffected, suggesting that HSCT is a better treatment option for patients with higher ARHGAP43 (SH3BP1) expression. KEGG and GSEA analyses revealed that high-expression ARHGAP43 (SH3BP1) was related to inflammation and immune response. Additionally, down-regulation of ARHGAP43 (SH3BP1) expression inhibited AML cell proliferation. CONCLUSION These findings highlight the clinical potential of ARHGAP43 (SH3BP1) as a novel biomarker of AML, with higher levels indicating a poor prognosis.
Collapse
Affiliation(s)
- Li Yang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Qiang Xu
- Department of Orthopedics and Joint Surgery, Qijiang District People's Hospital, Chongqing, 401420, China
| | - Junnan Li
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
3
|
Drouillard D, Halyko M, Cinquegrani E, McAllister D, Peterson FC, Marchese A, Dwinell MB. CXCL12 chemokine dimer signaling modulates acute myelogenous leukemia cell migration through altered receptor internalization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.26.609725. [PMID: 39253415 PMCID: PMC11383031 DOI: 10.1101/2024.08.26.609725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Acute myeloid leukemia (AML) is a malignancy of immature myeloid blast cells with stem-like and chemoresistant cells being retained in the bone marrow through CXCL12-CXCR4 signaling. Current CXCR4 inhibitors mobilize AML cells into the bloodstream where they become more chemosensitive have failed to improve patient survival, likely reflecting persistent receptor localization on target cells. Here we characterize the signaling properties of CXCL12-locked dimer (CXCL12-LD), a bioengineered variant of the dimeric CXCL12 structure. CXCL12-LD binding resulted in lower levels of G protein, β-arrestin, and intracellular calcium mobilization, consistent with the locked dimer being a partial agonist of CXCR4. Further, CXCL12-LD failed to induce chemotaxis in AML cells. Despite these partial agonist properties, CXCL12-LD increased CXCR4 internalization compared to wildtype and locked-monomer forms of CXCL12. Analysis of a previously published AML transcriptomic data showed CXCR4 positive AML cells co-express genes involved in chemoresistance and maintenance of a blast-like state. The CXCL12-LD partial agonist effectively mobilized stem cells into the bloodstream in mice suggesting a potential role for their use in targeting CXCR4. Together, our results suggest that enhanced internalization by CXCL12-LD partial agonist signaling can avoid pharmacodynamic tolerance and may identify new avenues to better target GPCRs.
Collapse
Affiliation(s)
- Donovan Drouillard
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee WI, USA
- Center for Immunology, Medical College of Wisconsin, Milwaukee WI, USA
| | - Michael Halyko
- Center for Immunology, Medical College of Wisconsin, Milwaukee WI, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee WI, USA
| | | | - Donna McAllister
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee WI, USA
- Center for Immunology, Medical College of Wisconsin, Milwaukee WI, USA
| | | | - Adriano Marchese
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee WI, USA
| | - Michael B. Dwinell
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee WI, USA
- Center for Immunology, Medical College of Wisconsin, Milwaukee WI, USA
- Department of Surgery, Medical College of Wisconsin, Milwaukee WI, USA
| |
Collapse
|
4
|
Janani G, Girigoswami A, Girigoswami K. Advantages of nanomedicine over the conventional treatment in Acute myeloid leukemia. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024; 35:415-441. [PMID: 38113194 DOI: 10.1080/09205063.2023.2294541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 12/08/2023] [Indexed: 12/21/2023]
Abstract
Leukemia is a cancer of blood cells that mainly affects the white blood cells. In acute myeloid leukemia (AML) sudden growth of cancerous cells occurs in blood and bone marrow, and it disrupts normal blood cell production. Most patients are asymptomatic, but it spreads rapidly and can become fatal if left untreated. AML is the prevalent form of leukemia in children. Risk factors of AML include chemical exposure, radiation, genetics, etc. Conventional diagnostic methods of AML are complete blood count tests and bone marrow aspiration, while conventional treatment methods involve chemotherapy, radiation therapy, and bone marrow transplant. There is a risk of cancer cells spreading progressively to the other organs if left untreated, and hence, early diagnosis is required. The conventional diagnostic methods are time- consuming and have drawbacks like harmful side effects and recurrence of the disease. To overcome these difficulties, nanoparticles are employed in treating and diagnosing AML. These nanoparticles can be surface- modified and can be used against cancer cells. Due to their enhanced permeability effect and high surface-to-volume ratio they will be able to reach the tumour site which cannot be reached by traditional drugs. This review article talks about how nanotechnology is more advantageous over the traditional methods in the treatment and diagnosis of AML.
Collapse
Affiliation(s)
- Gopalarethinam Janani
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chennai, Tamil Nadu, India
| | - Agnishwar Girigoswami
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chennai, Tamil Nadu, India
| | - Koyeli Girigoswami
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chennai, Tamil Nadu, India
| |
Collapse
|
5
|
Ogana HA, Hurwitz S, Wei N, Lee E, Morris K, Parikh K, Kim YM. Targeting integrins in drug-resistant acute myeloid leukaemia. Br J Pharmacol 2024; 181:295-316. [PMID: 37258706 DOI: 10.1111/bph.16149] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 04/14/2023] [Accepted: 05/10/2023] [Indexed: 06/02/2023] Open
Abstract
Acute myeloid leukaemia (AML) continues to have a poor prognosis, warranting new therapeutic strategies. The bone marrow (BM) microenvironment consists of niches that interact with not only normal haematopoietic stem cells (HSC) but also leukaemia cells like AML. There are many adhesion molecules in the BM microenvironment; therein, integrins have been of central interest. AML cells express integrins that bind to ligands in the microenvironment, enabling adhesion of leukaemia cells in the microenvironment, thereby initiating intracellular signalling pathways that are associated with cell migration, cell proliferation, survival, and drug resistance that has been described to mediate cell adhesion-mediated drug resistance (CAM-DR). Identifying and targeting integrins in AML to interrupt interactions with the microenvironment have been pursued as a strategy to overcome CAM-DR. Here, we focus on the BM microenvironment and review the role of integrins in CAM-DR of AML and discuss integrin-targeting strategies. LINKED ARTICLES: This article is part of a themed issue on Cancer Microenvironment and Pharmacological Interventions. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.2/issuetoc.
Collapse
Affiliation(s)
- Heather A Ogana
- Children's Hospital Los Angeles, Department of Pediatrics, Division of Hematology and Oncology, Cancer and Blood Disease Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Samantha Hurwitz
- Children's Hospital Los Angeles, Department of Pediatrics, Division of Hematology and Oncology, Cancer and Blood Disease Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Nathan Wei
- Children's Hospital Los Angeles, Department of Pediatrics, Division of Hematology and Oncology, Cancer and Blood Disease Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Eliana Lee
- Children's Hospital Los Angeles, Department of Pediatrics, Division of Hematology and Oncology, Cancer and Blood Disease Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Kayla Morris
- Children's Hospital Los Angeles, Department of Pediatrics, Division of Hematology and Oncology, Cancer and Blood Disease Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Karina Parikh
- Children's Hospital Los Angeles, Department of Pediatrics, Division of Hematology and Oncology, Cancer and Blood Disease Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Yong-Mi Kim
- Children's Hospital Los Angeles, Department of Pediatrics, Division of Hematology and Oncology, Cancer and Blood Disease Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
6
|
Pendse S, Chavan S, Kale V, Vaidya A. A comprehensive analysis of cell-autonomous and non-cell-autonomous regulation of myeloid leukemic cells: The prospect of developing novel niche-targeting therapies. Cell Biol Int 2023; 47:1667-1683. [PMID: 37554060 DOI: 10.1002/cbin.12078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/12/2023] [Accepted: 07/21/2023] [Indexed: 08/10/2023]
Abstract
Leukemic cells (LCs) arise from the hematopoietic stem/and progenitor cells (HSCs/HSPCs) and utilize cues from the bone marrow microenvironment (BMM) for their regulation in the same way as their normal HSC counterparts. Mesenchymal stromal cells (MSCs), a vital component of the BMM promote leukemogenesis by creating a protective and immune-tolerant microenvironment that can support the survival of LCs, helping them escape chemotherapy, thereby resulting in the relapse of leukemia. Conversely, MSCs also induce apoptosis in the LCs and inhibit their proliferation by interfering with their self-renewal potential. This review discusses the work done so far on cell-autonomous (intrinsic) and MSCs-mediated non-cell-autonomous (extrinsic) regulation of myeloid leukemia with a special focus on the need to investigate the extrinsic regulation of myeloid leukemia to understand the contrasting role of MSCs in leukemogenesis. These mechanisms could be exploited to formulate novel therapeutic strategies that specifically target the leukemic microenvironment.
Collapse
Affiliation(s)
- Shalmali Pendse
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, Maharashtra, India
- Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune, Maharashtra, India
| | - Sayali Chavan
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, Maharashtra, India
- Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune, Maharashtra, India
| | - Vaijayanti Kale
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, Maharashtra, India
- Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune, Maharashtra, India
| | - Anuradha Vaidya
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, Maharashtra, India
- Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune, Maharashtra, India
| |
Collapse
|
7
|
Bakhtiyari M, Liaghat M, Aziziyan F, Shapourian H, Yahyazadeh S, Alipour M, Shahveh S, Maleki-Sheikhabadi F, Halimi H, Forghaniesfidvajani R, Zalpoor H, Nabi-Afjadi M, Pornour M. The role of bone marrow microenvironment (BMM) cells in acute myeloid leukemia (AML) progression: immune checkpoints, metabolic checkpoints, and signaling pathways. Cell Commun Signal 2023; 21:252. [PMID: 37735675 PMCID: PMC10512514 DOI: 10.1186/s12964-023-01282-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/17/2023] [Indexed: 09/23/2023] Open
Abstract
Acute myeloid leukemia (AML) comprises a multifarious and heterogeneous array of illnesses characterized by the anomalous proliferation of myeloid cells in the bone marrow microenvironment (BMM). The BMM plays a pivotal role in promoting AML progression, angiogenesis, and metastasis. The immune checkpoints (ICs) and metabolic processes are the key players in this process. In this review, we delineate the metabolic and immune checkpoint characteristics of the AML BMM, with a focus on the roles of BMM cells e.g. tumor-associated macrophages, natural killer cells, dendritic cells, metabolic profiles and related signaling pathways. We also discuss the signaling pathways stimulated in AML cells by BMM factors that lead to AML progression. We then delve into the roles of immune checkpoints in AML angiogenesis, metastasis, and cell proliferation, including co-stimulatory and inhibitory ICs. Lastly, we discuss the potential therapeutic approaches and future directions for AML treatment, emphasizing the potential of targeting metabolic and immune checkpoints in AML BMM as prognostic and therapeutic targets. In conclusion, the modulation of these processes through the use of directed drugs opens up new promising avenues in combating AML. Thereby, a comprehensive elucidation of the significance of these AML BMM cells' metabolic and immune checkpoints and signaling pathways on leukemic cells can be undertaken in the future investigations. Additionally, these checkpoints and cells should be considered plausible multi-targeted therapies for AML in combination with other conventional treatments in AML. Video Abstract.
Collapse
Affiliation(s)
- Maryam Bakhtiyari
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Mahsa Liaghat
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Department of Medical Laboratory Sciences, Faculty of Medical Sciences, Kazerun Branch, Islamic Azad University, Kazerun, Iran
| | - Fatemeh Aziziyan
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hooriyeh Shapourian
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sheida Yahyazadeh
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maedeh Alipour
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Shaghayegh Shahveh
- American Association of Naturopath Physician (AANP), Washington, DC, USA
| | - Fahimeh Maleki-Sheikhabadi
- Department of Hematology and Blood Banking, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Halimi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Razieh Forghaniesfidvajani
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Hamidreza Zalpoor
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran.
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Majid Pornour
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, MD, USA.
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, Maryland, USA.
| |
Collapse
|
8
|
Zhao Y, Guo R, Cao X, Zhang Y, Sun R, Lu W, Zhao M. Role of chemokines in T-cell acute lymphoblastic Leukemia: From pathogenesis to therapeutic options. Int Immunopharmacol 2023; 121:110396. [PMID: 37295031 DOI: 10.1016/j.intimp.2023.110396] [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: 03/14/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/11/2023]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is a highly heterogeneous and aggressive subtype of hematologic malignancy, with limited therapeutic options due to the complexity of its pathogenesis. Although high-dose chemotherapy and allogeneic hematopoietic stem cell transplantation have improved outcomes for T-ALL patients, there remains an urgent need for novel treatments in cases of refractory or relapsed disease. Recent research has demonstrated the potential of targeted therapies aimed at specific molecular pathways to improve patient outcomes. Chemokine-related signals, both upstream and downstream, modulate the composition of distinct tumor microenvironments, thereby regulating a multitude of intricate cellular processes such as proliferation, migration, invasion and homing. Furthermore, the progress in research has made significant contributions to precision medicine by targeting chemokine-related pathways. This review article summarizes the crucial roles of chemokines and their receptors in T-ALL pathogenesis. Moreover, it explores the advantages and disadvantages of current and potential therapeutic options that target chemokine axes, including small molecule antagonists, monoclonal antibodies, and chimeric antigen receptor T-cells.
Collapse
Affiliation(s)
- YiFan Zhao
- First Center Clinic College of Tianjin Medical University, Tianjin 300192, China
| | - RuiTing Guo
- First Center Clinic College of Tianjin Medical University, Tianjin 300192, China
| | - XinPing Cao
- First Center Clinic College of Tianjin Medical University, Tianjin 300192, China
| | - Yi Zhang
- First Center Clinic College of Tianjin Medical University, Tianjin 300192, China
| | - Rui Sun
- School of Medicine, Nankai University, Tianjin 300192, China
| | - WenYi Lu
- Department of Hematology, Tianjin First Central Hospital, Tianjin 300192, China
| | - MingFeng Zhao
- Department of Hematology, Tianjin First Central Hospital, Tianjin 300192, China.
| |
Collapse
|
9
|
Anderson NR, Sheth V, Li H, Harris MW, Qiu S, Crossman DK, Kumar H, Agarwal P, Nagasawa T, Paterson AJ, Welner RS, Bhatia R. Microenvironmental CXCL12 deletion enhances Flt3-ITD acute myeloid leukemia stem cell response to therapy by reducing p38 MAPK signaling. Leukemia 2023; 37:560-570. [PMID: 36550214 PMCID: PMC10750268 DOI: 10.1038/s41375-022-01798-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 12/07/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022]
Abstract
Fms-like tyrosine kinase 3 (Flt3) tyrosine kinase inhibitors (Flt3-TKI) have improved outcomes for patients with Flt3-mutated acute myeloid leukemia (AML) but are limited by resistance and relapse, indicating persistence of leukemia stem cells (LSC). Here utilizing a Flt3-internal tandem duplication (Flt3-ITD) and Tet2-deleted AML genetic mouse model we determined that FLT3-ITD AML LSC were enriched within the primitive ST-HSC population. FLT3-ITD LSC showed increased expression of the CXCL12 receptor CXCR4. CXCL12-abundant reticular (CAR) cells were increased in Flt3-ITD AML marrow. CXCL12 deletion from the microenvironment enhanced targeting of AML cells by Flt3-TKI plus chemotherapy treatment, including enhanced LSC targeting. Both treatment and CXCL12 deletion partially reduced p38 mitogen-activated protein kinase (p38) signaling in AML cells and further reduction was seen after treatment in CXCL12 deleted mice. p38 inhibition reduced CXCL12-dependent and -independent maintenance of both murine and human Flt3-ITD AML LSC by MSC and enhanced their sensitivity to treatment. p38 inhibition in combination with chemotherapy plus TKI treatment leads to greater depletion of Flt3-ITD AML LSC compared with CXCL12 deletion. Our studies support roles for CXCL12 and p38 signaling in microenvironmental protection of AML LSC and provide a rationale for inhibiting p38 signaling to enhance Flt3-ITD AML targeting.
Collapse
Affiliation(s)
- Nicholas R Anderson
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Vipul Sheth
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hui Li
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mason W Harris
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shaowei Qiu
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
| | - David K Crossman
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Harish Kumar
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Puneet Agarwal
- Division of Experimental Hematology & Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Takashi Nagasawa
- Laboratory of Stem Cell Biology & Developmental Immunology, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Andrew J Paterson
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Robert S Welner
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ravi Bhatia
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
10
|
T22-PE24-H6 Nanotoxin Selectively Kills CXCR4-High Expressing AML Patient Cells In Vitro and Potently Blocks Dissemination In Vivo. Pharmaceutics 2023; 15:pharmaceutics15030727. [PMID: 36986589 PMCID: PMC10054149 DOI: 10.3390/pharmaceutics15030727] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/15/2023] [Accepted: 02/20/2023] [Indexed: 02/24/2023] Open
Abstract
Despite advances in the development of targeted therapies for acute myeloid leukemia (AML), most patients relapse. For that reason, it is still necessary to develop novel therapies that improve treatment effectiveness and overcome drug resistance. We developed T22-PE24-H6, a protein nanoparticle that contains the exotoxin A from the bacterium Pseudomonas aeruginosa and is able to specifically deliver this cytotoxic domain to CXCR4+ leukemic cells. Next, we evaluated the selective delivery and antitumor activity of T22-PE24-H6 in CXCR4+ AML cell lines and BM samples from AML patients. Moreover, we assessed the in vivo antitumor effect of this nanotoxin in a disseminated mouse model generated from CXCR4+ AML cells. T22-PE24-H6 showed a potent, CXCR4-dependent antineoplastic effect in vitro in the MONO-MAC-6 AML cell line. In addition, mice treated with nanotoxins in daily doses reduced the dissemination of CXCR4+ AML cells compared to buffer-treated mice, as shown by the significant decrease in BLI signaling. Furthermore, we did not observe any sign of toxicity or changes in mouse body weight, biochemical parameters, or histopathology in normal tissues. Finally, T22-PE24-H6 exhibited a significant inhibition of cell viability in CXCR4high AML patient samples but showed no activity in CXCR4low samples. These data strongly support the use of T22-PE24-H6 therapy to benefit high-CXCR4-expressing AML patients.
Collapse
|
11
|
Gupta DS, Gadi V, Kaur G, Chintamaneni M, Tuli HS, Ramniwas S, Sethi G. Resveratrol and Its Role in the Management of B-Cell Malignancies-A Recent Update. Biomedicines 2023; 11:221. [PMID: 36672729 PMCID: PMC9855921 DOI: 10.3390/biomedicines11010221] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/11/2023] [Accepted: 01/11/2023] [Indexed: 01/17/2023] Open
Abstract
The growing incidence of B cell malignancies globally has prompted research on the pharmacological properties of phytoconstituents in cancer management. Resveratrol, a polyphenolic stilbenoid widely found in nature, has been explored for its anti-inflammatory and antioxidant properties, and promising results from different pre-clinical studies have indicated its potential for management of B cell malignancies. However, these claims must be substantiated by a greater number of clinical trials in diverse populations, in order to establish its safety and efficacy profile. In addition to this, there is a need to explore nanodelivery of this agent, owing to its poor solubility, which in turn may impact its bioavailability. This review aims to offer an overview of the occurrence and pathogenesis of B cell malignancies with a special focus on the inflammatory pathways involved, the mechanism of actions of resveratrol and its pharmacokinetic profile, results from pre-clinical and clinical studies, as well as an overview of the marketed formulations. The authors have also presented their opinion on the various challenges associated with the clinical development of resveratrol and future perspectives regarding therapeutic applications of this agent.
Collapse
Affiliation(s)
- Dhruv Sanjay Gupta
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM’S NMIMS, Vile Parle-West, Mumbai 400056, India
| | - Vaishnavi Gadi
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM’S NMIMS, Vile Parle-West, Mumbai 400056, India
| | - Ginpreet Kaur
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM’S NMIMS, Vile Parle-West, Mumbai 400056, India
| | - Meena Chintamaneni
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM’S NMIMS, Vile Parle-West, Mumbai 400056, India
| | - Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala 133207, India
| | - Seema Ramniwas
- University Centre for Research and Development, University Institute of Pharmaceutical Sciences, Chandigarh University, Gharuan, Mohali 140413, India
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| |
Collapse
|
12
|
Bouligny IM, Maher KR, Grant S. Mechanisms of myeloid leukemogenesis: Current perspectives and therapeutic objectives. Blood Rev 2023; 57:100996. [PMID: 35989139 PMCID: PMC10693933 DOI: 10.1016/j.blre.2022.100996] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 01/28/2023]
Abstract
Acute myeloid leukemia (AML) is a heterogeneous hematopoietic neoplasm which results in clonal proliferation of abnormally differentiated hematopoietic cells. In this review, mechanisms contributing to myeloid leukemogenesis are summarized, highlighting aberrations of epigenetics, transcription factors, signal transduction, cell cycling, and the bone marrow microenvironment. The mechanisms contributing to AML are detailed to spotlight recent findings that convey clinical impact. The applications of current and prospective therapeutic targets are accentuated in addition to reviews of treatment paradigms stratified for each characteristic molecular lesion - with a focus on exploring novel treatment approaches and combinations to improve outcomes in AML.
Collapse
Affiliation(s)
- Ian M Bouligny
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| | - Keri R Maher
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| | - Steven Grant
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
13
|
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: 23] [Impact Index Per Article: 7.7] [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
|
14
|
Díaz-Beyá M, García-Fortes M, Valls R, Artigas L, Gómez-Casares MT, Montesinos P, Sánchez-Guijo F, Coma M, Vendranes M, Martínez-López J. A Systems Biology- and Machine Learning-Based Study to Unravel Potential Therapeutic Mechanisms of Midostaurin as a Multitarget Therapy on FLT3-Mutated AML. BIOMEDINFORMATICS 2022; 2:375-397. [DOI: 10.3390/biomedinformatics2030024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Acute myeloid leukemia (AML), a hematologic malignancy that results in bone marrow failure, is the most common acute leukemia in adults. The presence of FMS-related tyrosine kinase 3 (FLT3) mutations is associated with a poor prognosis, making the evaluation of FLT3-inhibitors an imperative goal in clinical trials. Midostaurin was the first FLT3-inhibitor approved by the FDA and EMA for the treatment of FLT3-mutated AML, and it showed a significant improvement in overall survival for newly diagnosed patients treated with midostaurin, in combination with standard chemotherapy (RATIFY study). The main interest of midostaurin has been the FLT3-specific inhibition, but little is known about its role as a multikinase inhibitor and whether it may be used in relapse and maintenance therapy. Here, we used systems biology- and machine learning-based approaches to deepen the potential benefits of the multitarget activity of midostaurin and to better understand its anti-leukemic effect on FLT3-mutated AML. The resulting in silico study revealed that the multikinase activity of midostaurin may play a role in the treatment’s efficacy. Additionally, we propose a series of molecular mechanisms that support a potential benefit of midostaurin as a maintenance therapy in FLT3-mutated AML, by regulating the microenvironment. The obtained results are backed up using independent gene expression data.
Collapse
Affiliation(s)
- Marina Díaz-Beyá
- Department of Hematology, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Myeloid Neoplasms Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Spain
| | - María García-Fortes
- Hematology Department, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
| | - Raquel Valls
- Molecular Health Department, Anaxomics Biotech SL, 08007 Barcelona, Spain
| | - Laura Artigas
- Molecular Health Department, Anaxomics Biotech SL, 08007 Barcelona, Spain
| | - Mª Teresa Gómez-Casares
- Hematology Service, Hospital Universitario Insular Materno-Infantil, 35016 Las Palmas de Gran Canaria, Spain
| | - Pau Montesinos
- Departament of Hematology, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
| | - Fermín Sánchez-Guijo
- Cancer Research Center (CIC), Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, Universidad de Salamanca, 37007 Salamanca, Spain
| | - Mireia Coma
- Molecular Health Department, Anaxomics Biotech SL, 08007 Barcelona, Spain
| | | | - Joaquín Martínez-López
- Hospital 12 de Octubre. Universidad Complutense. CNIO. Centro de Investigación Biomédica en Red-Oncología (CIBERONC), Instituto Carlos III, 28041 Madrid, Spain
| |
Collapse
|
15
|
Li H, Wu M, Zhao X. Role of chemokine systems in cancer and inflammatory diseases. MedComm (Beijing) 2022; 3:e147. [PMID: 35702353 PMCID: PMC9175564 DOI: 10.1002/mco2.147] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 12/12/2022] Open
Abstract
Chemokines are a large family of small secreted proteins that have fundamental roles in organ development, normal physiology, and immune responses upon binding to their corresponding receptors. The primary functions of chemokines are to coordinate and recruit immune cells to and from tissues and to participate in regulating interactions between immune cells. In addition to the generally recognized antimicrobial immunity, the chemokine/chemokine receptor axis also exerts a tumorigenic function in many different cancer models and is involved in the formation of immunosuppressive and protective tumor microenvironment (TME), making them potential prognostic markers for various hematologic and solid tumors. In fact, apart from its vital role in tumors, almost all inflammatory diseases involve chemokines and their receptors in one way or another. Modulating the expression of chemokines and/or their corresponding receptors on tumor cells or immune cells provides the basis for the exploitation of new drugs for clinical evaluation in the treatment of related diseases. Here, we summarize recent advances of chemokine systems in protumor and antitumor immune responses and discuss the prevailing understanding of how the chemokine system operates in inflammatory diseases. In this review, we also emphatically highlight the complexity of the chemokine system and explore its potential to guide the treatment of cancer and inflammatory diseases.
Collapse
Affiliation(s)
- Hongyi Li
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of EducationWest China Second HospitalSichuan UniversityChengduChina
| | - Min Wu
- Department of Biomedical Sciences, School of Medicine and Health SciencesUniversity of North DakotaGrand ForksNorth DakotaUSA
| | - Xia Zhao
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of EducationWest China Second HospitalSichuan UniversityChengduChina
| |
Collapse
|
16
|
One-Chip Isolation of Drug-Resistant Acute Myeloid Leukemia Cells with CXCR4-Targeted Magnetic Fluorescent Nanoprobes. NANOMATERIALS 2022; 12:nano12101711. [PMID: 35630929 PMCID: PMC9142899 DOI: 10.3390/nano12101711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 05/12/2022] [Accepted: 05/16/2022] [Indexed: 11/17/2022]
Abstract
Drug resistance and relapse lead to high mortality in acute myeloid leukemia, and studies have shown that CXCR4 overexpression is highly correlated with poor prognosis and drug resistance in leukemia cells. Isolation and detection of AML cells with CXCR4 overexpression will be crucial to the treatment of AML. In this paper, magnetic nanoparticles were firstly prepared successfully by high-temperature thermal decomposition method, and then characterized by TEM, VSM and DLS. Subsequently CXCR4-targeted magnetic fluorescent nanoprobes conjugated with antibody 12G5 were constructed by stepwise coupling. In cell experiments, the obtained probes demonstrated excellent targeting efficacy to CXCR4 overexpressed AML cells HL-60. In addition, HL-60 cells labelled with the magnetic probes can be magnetic isolated successfully in one microfluidics chip, with efficiency of 82.92 ± 7.03%. Overall, this method utilizes the superiority of superparamagnetic nanomaterials and microfluidic technology to achieve the enrichment and capture of drug-resistant cells in a microfluidic chip, providing a new idea for the isolation and detective of drug-resistant acute myeloid leukemia cells.
Collapse
|
17
|
Leukemic Stem Cells as a Target for Eliminating Acute Myeloid Leukemia: Gaps in Translational Research. Crit Rev Oncol Hematol 2022; 175:103710. [DOI: 10.1016/j.critrevonc.2022.103710] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 04/29/2022] [Accepted: 05/11/2022] [Indexed: 12/26/2022] Open
|
18
|
Ruiz-Aparicio PF, Vernot JP. Bone Marrow Aging and the Leukaemia-Induced Senescence of Mesenchymal Stem/Stromal Cells: Exploring Similarities. J Pers Med 2022; 12:jpm12050716. [PMID: 35629139 PMCID: PMC9147878 DOI: 10.3390/jpm12050716] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/20/2022] [Accepted: 04/27/2022] [Indexed: 12/17/2022] Open
Abstract
Bone marrow aging is associated with multiple cellular dysfunctions, including perturbed haematopoiesis, the propensity to haematological transformation, and the maintenance of leukaemia. It has been shown that instructive signals from different leukemic cells are delivered to stromal cells to remodel the bone marrow into a supportive leukemic niche. In particular, cellular senescence, a physiological program with both beneficial and deleterious effects on the health of the organisms, may be responsible for the increased incidence of haematological malignancies in the elderly and for the survival of diverse leukemic cells. Here, we will review the connection between BM aging and cellular senescence and the role that these processes play in leukaemia progression. Specifically, we discuss the role of mesenchymal stem cells as a central component of the supportive niche. Due to the specificity of the genetic defects present in leukaemia, one would think that bone marrow alterations would also have particular changes, making it difficult to envisage a shared therapeutic use. We have tried to summarize the coincident features present in BM stromal cells during aging and senescence and in two different leukaemias, acute myeloid leukaemia, with high frequency in the elderly, and B-acute lymphoblastic leukaemia, mainly a childhood disease. We propose that mesenchymal stem cells are similarly affected in these different leukaemias, and that the changes that we observed in terms of cellular function, redox balance, genetics and epigenetics, soluble factor repertoire and stemness are equivalent to those occurring during BM aging and cellular senescence. These coincident features may be used to explore strategies useful to treat various haematological malignancies.
Collapse
Affiliation(s)
- Paola Fernanda Ruiz-Aparicio
- Grupo de Investigación Fisiología Celular y Molecular, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá 111321, Colombia;
| | - Jean-Paul Vernot
- Grupo de Investigación Fisiología Celular y Molecular, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá 111321, Colombia;
- Instituto de Investigaciones Biomédicas, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá 111321, Colombia
- Correspondence:
| |
Collapse
|
19
|
Mehrpouri M. The contributory roles of the CXCL12/CXCR4/CXCR7 axis in normal and malignant hematopoiesis: A possible therapeutic target in hematologic malignancies. Eur J Pharmacol 2022; 920:174831. [PMID: 35183534 DOI: 10.1016/j.ejphar.2022.174831] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 11/03/2022]
|
20
|
Almohsen F, Al-Rubaie HA, Habib MA, Nasr SA, Perni R, Al-Quraishi L. Circulating miR-126-3p and miR-423-5p Expression in de novo Adult Acute Myeloid Leukemia: Correlations with Response to Induction Therapy and the 2-Year Overall Survival. J Blood Med 2022; 13:83-92. [PMID: 35210895 PMCID: PMC8863343 DOI: 10.2147/jbm.s347397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/20/2022] [Indexed: 12/02/2022] Open
Abstract
Background Purpose Patients and Methods Results Conclusion
Collapse
Affiliation(s)
- Faez Almohsen
- College of Medicine, University of Baghdad, Baghdad, Iraq
- Correspondence: Faez Almohsen, College of Medicine, University of Baghdad, Baghdad, Iraq, Tel +964 7902834062, Email
| | | | - Manal A Habib
- College of Medicine, University of Baghdad, Baghdad, Iraq
| | - Sherif A Nasr
- siParadigm Diagnostic Informatics, New Jersey, NJ, USA
| | | | | |
Collapse
|
21
|
Ganesan S, Mathews V, Vyas N. Microenvironment and drug resistance in acute myeloid leukemia: Do we know enough? Int J Cancer 2021; 150:1401-1411. [PMID: 34921734 DOI: 10.1002/ijc.33908] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 12/09/2021] [Accepted: 12/10/2021] [Indexed: 12/18/2022]
Abstract
Acute myeloid leukemia (AMLs), as the name suggests, often develop suddenly and are very progressive forms of cancer. Unlike in acute promyelocytic leukemia, a subtype of AML, the outcomes in most other AMLs remain poor. This is mainly attributed to the acquired drug resistance and lack of targeted therapy. Different studies across laboratories suggest that the cellular mechanisms to impart therapy resistance are often very dynamic and should be identified in a context-specific manner. Our review highlights the progress made so far in identifying the different cellular mechanisms of mutation-independent therapy resistance in AML. It reiterates that for more effective outcomes cancer therapies should acquire a more tailored approach where the protective interactions between the cancer cells and their niches are identified and targeted.
Collapse
Affiliation(s)
- Saravanan Ganesan
- Department of Haematology, Christian Medical College, Vellore, India
| | - Vikram Mathews
- Department of Haematology, Christian Medical College, Vellore, India
| | - Neha Vyas
- Division of Molecular Medicine, St. John's Research Institute, SJNAHS, Bengaluru, India
| |
Collapse
|
22
|
Mitochondria and the Tumour Microenvironment in Blood Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1329:181-203. [PMID: 34664240 DOI: 10.1007/978-3-030-73119-9_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
The bone marrow (BM) is a complex organ located within the cavities of bones. The main function of the BM is to produce all the blood cells required for a normal healthy blood system. As with any major organ, many diseases can arise from errors in bone marrow function, including non-malignant disorders such as anaemia and malignant disorders such as leukaemias. This article will explore the role of the bone marrow, in normal and diseased haematopoiesis, with an emphasis on the requirement for intercellular mitochondrial transfer in leukaemia.
Collapse
|
23
|
Bolandi SM, Pakjoo M, Beigi P, Kiani M, Allahgholipour A, Goudarzi N, Khorashad JS, Eiring AM. A Role for the Bone Marrow Microenvironment in Drug Resistance of Acute Myeloid Leukemia. Cells 2021; 10:2833. [PMID: 34831055 PMCID: PMC8616250 DOI: 10.3390/cells10112833] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 02/08/2023] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease with a poor prognosis and remarkable resistance to chemotherapeutic agents. Understanding resistance mechanisms against currently available drugs helps to recognize the therapeutic obstacles. Various mechanisms of resistance to chemotherapy or targeted inhibitors have been described for AML cells, including a role for the bone marrow niche in both the initiation and persistence of the disease, and in drug resistance of the leukemic stem cell (LSC) population. The BM niche supports LSC survival through direct and indirect interactions among the stromal cells, hematopoietic stem/progenitor cells, and leukemic cells. Additionally, the BM niche mediates changes in metabolic and signal pathway activation due to the acquisition of new mutations or selection and expansion of a minor clone. This review briefly discusses the role of the BM microenvironment and metabolic pathways in resistance to therapy, as discovered through AML clinical studies or cell line and animal models.
Collapse
Affiliation(s)
- Seyed Mohammadreza Bolandi
- Department of Immunology, Razi Vaccine and Sera Research Institute, Karaj, Iran; (S.M.B.); (N.G.)
- Department of Pharmacology, Karaj Branch, Islamic Azad University, Karaj, Iran; (M.K.); (A.A.)
| | - Mahdi Pakjoo
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; (M.P.); (P.B.)
| | - Peyman Beigi
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; (M.P.); (P.B.)
| | - Mohammad Kiani
- Department of Pharmacology, Karaj Branch, Islamic Azad University, Karaj, Iran; (M.K.); (A.A.)
| | - Ali Allahgholipour
- Department of Pharmacology, Karaj Branch, Islamic Azad University, Karaj, Iran; (M.K.); (A.A.)
| | - Negar Goudarzi
- Department of Immunology, Razi Vaccine and Sera Research Institute, Karaj, Iran; (S.M.B.); (N.G.)
| | - Jamshid S. Khorashad
- Centre for Haematology, Hammersmith Hospital, Imperial College London, London W12 0HS, UK;
| | - Anna M. Eiring
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center at El Paso, El Paso, TX 79905, USA
| |
Collapse
|
24
|
Stetson LC, Balasubramanian D, Ribeiro SP, Stefan T, Gupta K, Xu X, Fourati S, Roe A, Jackson Z, Schauner R, Sharma A, Tamilselvan B, Li S, de Lima M, Hwang TH, Balderas R, Saunthararajah Y, Maciejewski J, LaFramboise T, Barnholtz-Sloan JS, Sekaly RP, Wald DN. Single cell RNA sequencing of AML initiating cells reveals RNA-based evolution during disease progression. Leukemia 2021; 35:2799-2812. [PMID: 34244611 PMCID: PMC8807029 DOI: 10.1038/s41375-021-01338-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/19/2021] [Accepted: 06/25/2021] [Indexed: 02/06/2023]
Abstract
The prognosis of most patients with AML is poor due to frequent disease relapse. The cause of relapse is thought to be from the persistence of leukemia initiating cells (LIC's) following treatment. Here we assessed RNA based changes in LICs from matched patient diagnosis and relapse samples using single-cell RNA sequencing. Previous studies on AML progression have focused on genetic changes at the DNA mutation level mostly in bulk AML cells and demonstrated the existence of DNA clonal evolution. Here we identified in LICs that the phenomenon of RNA clonal evolution occurs during AML progression. Despite the presence of vast transcriptional heterogeneity at the single cell level, pathway analysis identified common signaling networks involving metabolism, apoptosis and chemokine signaling that evolved during AML progression and become a signature of relapse samples. A subset of this gene signature was validated at the protein level in LICs by flow cytometry from an independent AML cohort and functional studies were performed to demonstrate co-targeting BCL2 and CXCR4 signaling may help overcome therapeutic challenges with AML heterogeneity. It is hoped this work will facilitate a greater understanding of AML relapse leading to improved prognostic biomarkers and therapeutic strategies to target LIC's.
Collapse
Affiliation(s)
- L C Stetson
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | | | | | - Tammy Stefan
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Kalpana Gupta
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Xuan Xu
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Slim Fourati
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Anne Roe
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Zachary Jackson
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Robert Schauner
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Ashish Sharma
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | | | - Samuel Li
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Marcos de Lima
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
- Department of Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Tae Hyun Hwang
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, USA
| | | | - Yogen Saunthararajah
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
- Department of Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, OH, USA
| | - Jaroslaw Maciejewski
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
- Department of Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, OH, USA
| | - Thomas LaFramboise
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Jill S Barnholtz-Sloan
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Rafick-Pierre Sekaly
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - David N Wald
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA.
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA.
- Department of Pathology, University Hospitals Cleveland Medical Center and Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA.
| |
Collapse
|
25
|
Yılmaz H, Toy HI, Marquardt S, Karakülah G, Küçük C, Kontou PI, Logotheti S, Pavlopoulou A. In Silico Methods for the Identification of Diagnostic and Favorable Prognostic Markers in Acute Myeloid Leukemia. Int J Mol Sci 2021; 22:ijms22179601. [PMID: 34502522 PMCID: PMC8431757 DOI: 10.3390/ijms22179601] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/13/2021] [Accepted: 08/20/2021] [Indexed: 12/13/2022] Open
Abstract
Acute myeloid leukemia (AML), the most common type of acute leukemia in adults, is mainly asymptomatic at early stages and progresses/recurs rapidly and frequently. These attributes necessitate the identification of biomarkers for timely diagnosis and accurate prognosis. In this study, differential gene expression analysis was performed on large-scale transcriptomics data of AML patients versus corresponding normal tissue. Weighted gene co-expression network analysis was conducted to construct networks of co-expressed genes, and detect gene modules. Finally, hub genes were identified from selected modules by applying network-based methods. This robust and integrative bioinformatics approach revealed a set of twenty-four genes, mainly related to cell cycle and immune response, the diagnostic significance of which was subsequently compared against two independent gene expression datasets. Furthermore, based on a recent notion suggesting that molecular characteristics of a few, unusual patients with exceptionally favorable survival can provide insights for improving the outcome of individuals with more typical disease trajectories, we defined groups of long-term survivors in AML patient cohorts and compared their transcriptomes versus the general population to infer favorable prognostic signatures. These findings could have potential applications in the clinical setting, in particular, in diagnosis and prognosis of AML.
Collapse
Affiliation(s)
- Hande Yılmaz
- Izmir Biomedicine and Genome Center, Balcova, 35340 Izmir, Turkey; (H.Y.); (H.I.T.); (G.K.); (C.K.)
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Balcova, 35340 Izmir, Turkey
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, 18057 Rostock, Germany;
| | - Halil Ibrahim Toy
- Izmir Biomedicine and Genome Center, Balcova, 35340 Izmir, Turkey; (H.Y.); (H.I.T.); (G.K.); (C.K.)
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Balcova, 35340 Izmir, Turkey
| | - Stephan Marquardt
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, 18057 Rostock, Germany;
| | - Gökhan Karakülah
- Izmir Biomedicine and Genome Center, Balcova, 35340 Izmir, Turkey; (H.Y.); (H.I.T.); (G.K.); (C.K.)
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Balcova, 35340 Izmir, Turkey
| | - Can Küçük
- Izmir Biomedicine and Genome Center, Balcova, 35340 Izmir, Turkey; (H.Y.); (H.I.T.); (G.K.); (C.K.)
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Balcova, 35340 Izmir, Turkey
- Department of Medical Biology, Faculty of Medicine, Dokuz Eylül University, Balcova, 35340 Izmir, Turkey
| | - Panagiota I. Kontou
- Department of Computer Science and Biomedical Informatics, University of Thessaly, 35131 Lamia, Greece;
| | - Stella Logotheti
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, 18057 Rostock, Germany;
- Correspondence: (S.L.); (A.P.)
| | - Athanasia Pavlopoulou
- Izmir Biomedicine and Genome Center, Balcova, 35340 Izmir, Turkey; (H.Y.); (H.I.T.); (G.K.); (C.K.)
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Balcova, 35340 Izmir, Turkey
- Correspondence: (S.L.); (A.P.)
| |
Collapse
|
26
|
Aref S, Ebrahim L, El-Ashwah S, El Agdar M, Ayed M. Relevance of plasma bone marrow Activin-A and CXCL-12 concentration levels as a biomarker in acute myeloid leukemia. Cancer Biomark 2021; 32:263-270. [PMID: 34092614 DOI: 10.3233/cbm-203171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Acute myeloid leukemia changes the bone marrow (BM) niche to support leukemia cells by modulating the stromal microenvironment. The aim is to assess Activin-A as a biomarker in acute myeloid leukemia (AML). METHODS The level of Activin-A and CXCL-12 protein concentration levels in the plasma of bone marrow aspirate samples of eighty AML patients at diagnosis, after induction and at relapse were determined by ELISA. RESULTS We found that Activin-A concentration levels was significantly up regulated in AML cases at diagnosis, and down regulated at complete remission and rise again at relapse (P< 0.001). In contrast; the CXCL-12 gene expression was significantly down regulated in AML cases at diagnosis; relapse, and up regulated after complete remission (P< 0.001). Multivariate analysis showed that high Activin-A levels at diagnosis is significant predictor of induction of remission response OR 1.006 (CI: 1.002-1.010) (P= 0.003); AML relapse OR 1.002 (CI: 1.0-1.004) (P= 0.043) as well as patients' outcome OR 1.33 (CI: 1.004-1.062) (P= 0.024). CONCLUSION Activin-A level at diagnosis is a new simple easily assessed biomarker that could predict AML patient's response to therapy as well as patient's outcome.
Collapse
Affiliation(s)
- Salah Aref
- Hematology Unit, Clinical Pathology Department, Mansoura Faculty of Medicine, Mansoura University, Egypt
| | - Lamiaa Ebrahim
- Hematology Unit, Clinical Pathology Department, Mansoura Faculty of Medicine, Mansoura University, Egypt
| | - Shaimaa El-Ashwah
- Hematology Unit, Mansoura University Oncology Center-Mansoura University, Egypt
| | - Mohamed El Agdar
- Hematology Unit, Clinical Pathology Department, Mansoura Faculty of Medicine, Mansoura University, Egypt
| | - Mohamed Ayed
- Hematology Unit, Clinical Pathology Department, Mansoura Faculty of Medicine, Mansoura University, Egypt
| |
Collapse
|
27
|
Pallarès V, Núñez Y, Sánchez-García L, Falgàs A, Serna N, Unzueta U, Gallardo A, Alba-Castellón L, Álamo P, Sierra J, Villaverde A, Vázquez E, Casanova I, Mangues R. Antineoplastic effect of a diphtheria toxin-based nanoparticle targeting acute myeloid leukemia cells overexpressing CXCR4. J Control Release 2021; 335:117-129. [PMID: 34004204 DOI: 10.1016/j.jconrel.2021.05.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 05/05/2021] [Accepted: 05/11/2021] [Indexed: 12/27/2022]
Abstract
Nanomedicine has opened an opportunity to improve current clinical practice by enhancing the selectivity in the delivery of antitumor drugs to specific cancer cells. These new strategies are able to bypass toxicity on normal cells increasing the effectiveness of current anticancer treatments. In acute myeloid leukemia (AML) current chemotherapy treatments generate a relevant toxic impact in normal cells and severe side effects or even patient death. In this study, we have designed a self-assembling protein nanoparticle, T22-DITOX-H6, which incorporates a ligand (T22) targeting CXCR4-overexpressing (CXCR4+) cells, and a potent cytotoxic diphtheria toxin domain. CXCR4 is overexpressed in AML leukemic cells and associates with poor prognosis, being, therefore, a relevant clinical target. We demonstrate here that T22-DITOX-H6 induces apoptosis in CXCR4+ leukemic cells through CXCR4-dependent internalization. In addition, repeated T22-DITOX-H6 treatment (10 μg/dose per 10 doses, intravenously injected) in a disseminated AML mouse model (NSG mice intravenously injected with THP-1-Luci cells, n = 10 per group) potently blocks the dissemination of AML cells in bone marrow, spleen and liver of treated mice, without inducing toxicity in healthy tissues. In conclusion, our strategy of selectively ablating CXCR4 positive leukemic cells by administering the T22-DITOX-H6 nanoparticle could be a promising treatment, especially in patients undergoing AML relapse after chemotherapy, in which leukemic cells overexpress CXCR4.
Collapse
Affiliation(s)
- Victor Pallarès
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Josep Carreras Research Institute, Barcelona, Spain; CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain
| | - Yáiza Núñez
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Josep Carreras Research Institute, Barcelona, Spain; CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain
| | - Laura Sánchez-García
- CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain; Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Barcelona, Spain; Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Aïda Falgàs
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Josep Carreras Research Institute, Barcelona, Spain; CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain
| | - Naroa Serna
- CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain; Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Barcelona, Spain; Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ugutz Unzueta
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Josep Carreras Research Institute, Barcelona, Spain; CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain; Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alberto Gallardo
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Department of Pathology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Lorena Alba-Castellón
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Josep Carreras Research Institute, Barcelona, Spain
| | - Patricia Álamo
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Josep Carreras Research Institute, Barcelona, Spain; CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain
| | - Jorge Sierra
- Josep Carreras Research Institute, Barcelona, Spain; Department of Hematology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Antonio Villaverde
- CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain; Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Barcelona, Spain; Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Esther Vázquez
- CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain; Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Barcelona, Spain; Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Isolda Casanova
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Josep Carreras Research Institute, Barcelona, Spain; CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain.
| | - Ramon Mangues
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Josep Carreras Research Institute, Barcelona, Spain; CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain.
| |
Collapse
|
28
|
O'Reilly E, Zeinabad HA, Szegezdi E. Hematopoietic versus leukemic stem cell quiescence: Challenges and therapeutic opportunities. Blood Rev 2021; 50:100850. [PMID: 34049731 DOI: 10.1016/j.blre.2021.100850] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 04/22/2021] [Accepted: 05/07/2021] [Indexed: 12/13/2022]
Abstract
Hematopoietic stem cells (HSC) are responsible for the production of mature blood cells. To ensure that the HSC pool does not get exhausted over the lifetime of an individual, most HSCs are in a state of quiescence with only a small proportion of HSCs dividing at any one time. HSC quiescence is carefully controlled by both intrinsic and extrinsic, niche-driven mechanisms. In acute myeloid leukemia (AML), the leukemic cells overtake the hematopoietic bone marrow niche where they acquire a quiescent state. These dormant AML cells are resistant to chemotherapeutics. Because they can re-establish the disease after therapy, they are often termed as quiescent leukemic stem cells (LSC) or leukemia-initiating cells. While advancements are being made to target particular driver mutations in AML, there is less focus on how to tackle the drug resistance of quiescent LSCs. This review summarises the current knowledge on the biochemical characteristics of quiescent HSCs and LSCs, the intracellular signaling pathways and the niche-driven mechanisms that control quiescence and the key differences between HSC- and LSC-quiescence that may be exploited for therapy.
Collapse
Affiliation(s)
- Eimear O'Reilly
- Apoptosis Research Centre, Department of Biochemistry, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Hojjat Alizadeh Zeinabad
- Apoptosis Research Centre, Department of Biochemistry, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Eva Szegezdi
- Apoptosis Research Centre, Department of Biochemistry, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland.
| |
Collapse
|
29
|
Importance of Altered Gene Expression of Metalloproteinases 2, 9, and 16 in Acute Myeloid Leukemia: Preliminary Study. JOURNAL OF ONCOLOGY 2021; 2021:6697975. [PMID: 34035811 PMCID: PMC8121570 DOI: 10.1155/2021/6697975] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 04/13/2021] [Accepted: 04/29/2021] [Indexed: 12/21/2022]
Abstract
Acute myeloid leukemia is a group of hematological neoplasms characterized by a heterogeneous course and high mortality. The important factor in the neoplastic process is metalloproteinases, proteolytic enzymes capable of degrading various components of the extracellular matrix, which take an active part in modifying the functioning of the cell, including transformation to cancer cell. They interact with numerous signaling pathways responsible for the process of cell growth, proliferation, or apoptosis. In the present study, changes in the expression of MMP2, MMP9, and MMP16 genes between patients with AML and people without cancer were examined. The impact of cytogenetic changes in neoplastic cells on the expression level of MMP2, MMP9, and MMP16 was also assessed, as well as the impact of the altered expression on the effectiveness of the first cycle of remission-inducing therapy. To evaluate the expression of all studied genes MMP2, MMP9, and MMP16, SYBR Green-based real-time PCR method was used; the reference gene was GAPDH. For two investigated genes MMP2 and MMP16, the lower expression level was observed in patients with AML when compared to healthy people. The MMP9 gene expression level did not differ between patients with AML and healthy individuals which may indicate a different regulation of gene expression in acute myeloid leukemia. However, no correlation was observed between the genes' expression of all tested metalloproteinases and the result of cytoreductive treatment or the presence of cytogenetic changes. The obtained results show that the expression of MMP2 and MMP16 genes is reduced while the expression of MMP9 is unchanged in patients with acute myeloid leukemia. This may indicate a different regulation of the expression of these genes, and possible disruptions in gene transcription or posttranscriptional mechanisms in the MMP2 and MMP16 genes, however, do not affect the level of MMP9 expression. Obtained results in AML patients are in contrary to various types of solid tumors where increased expression is usually observed.
Collapse
|
30
|
Royston D, Mead AJ, Psaila B. Application of Single-Cell Approaches to Study Myeloproliferative Neoplasm Biology. Hematol Oncol Clin North Am 2021; 35:279-293. [PMID: 33641869 PMCID: PMC7935666 DOI: 10.1016/j.hoc.2021.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Philadelphia-negative myeloproliferative neoplasms (MPNs) are an excellent tractable disease model of a number of aspects of human cancer biology, including genetic evolution, tissue-associated fibrosis, and cancer stem cells. In this review, we discuss recent insights into MPN biology gained from the application of a number of new single-cell technologies to study human disease, with a specific focus on single-cell genomics, single-cell transcriptomics, and digital pathology.
Collapse
Affiliation(s)
- Daniel Royston
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine and NIHR Biomedical Research Centre, University of Oxford, Headley Way, Oxford OX39DS, UK
| | - Adam J Mead
- Medical Research Council (MRC) Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, NIHR Biomedical Research Centre, University of Oxford, Headley Way, Oxford OX3 9DS, UK.
| | - Bethan Psaila
- Medical Research Council (MRC) Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, NIHR Biomedical Research Centre, University of Oxford, Headley Way, Oxford OX3 9DS, UK
| |
Collapse
|
31
|
Aladle DAAM, Ghannam MA, El-Ashwah S, Ghobrial FEI, Mortada MI. Association of SDF-1 Gene Polymorphism with Increased Risk of Acute Myeloid Leukemia Patients. Asian Pac J Cancer Prev 2021; 22:1035-1043. [PMID: 33906294 PMCID: PMC8325146 DOI: 10.31557/apjcp.2021.22.4.1035] [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: 07/03/2020] [Indexed: 11/25/2022] Open
Abstract
Background: Acute myeloid leukemia (AML) is a heterogenous group of disorders that emerge from the malignant transformation of hematopoietic stem cells. Chemokine stromal cell-derived factor 1(SDF-1) and its receptor CXC receptor 4 (CXCR4) has an essential role in dissemination of blast cells. Study aimed to detect CXCR4 expression and the SDF-1 (rs1801157) gene polymorphisms and correlate them with prognosis and outcome in AML patients. Subjects and Methods: The study was conducted on 60 de-novo AML patients, and 60 healthy controls. SDF-1 (rs1801157) gene polymorphisms were detected by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP), and CXCR4 expression was done using flow cytometry analysis. Results: SDF-1 dominant model (AG+AA) had higher risk AML (p 0.002). CXCR4 positive cases were associated significantly with toxic manifestations (p 0.019), lower CR rates (p 0.004), and unfavorable cytogenetics (p 0.027). Multivariate analysis showed that combined CXCR4positive with dominant SDF-1 considered as independent prognostic factor for shorter overall survival (OS) in AML patients (p 0.031). Conclusion: SDF-1 dominant model had a higher risk to develop AML, and CXCR4 positive expression predicts poor prognosis in AML patients and it could represent a targeted therapy in AML. In addition, CXCR4 could be easily integrated into the initial routine diagnostic work up of AML.
Collapse
Affiliation(s)
- Doaa Abd Allah M Aladle
- Hematology Unit, Department of Clinical Pathology, Mansoura Faculty of Medicine, Mansoura University, Egypt
| | - Mayada A Ghannam
- Hematology Unit, Department of Clinical Pathology, Mansoura Faculty of Medicine, Mansoura University, Egypt
| | - Shaimaa El-Ashwah
- Clinical Hematology Unit, Department of Internal Medicine, Oncology Center, Mansoura University, Mansoura, Egypt
| | - F E I Ghobrial
- Medical Oncology Unit, Department of Internal Medicine, Oncology Center, Faculty of Medicine, Mansoura University, Egypt
| | - Metwaly Ibrahim Mortada
- Hematology Unit, Department of Clinical Pathology, Mansoura Faculty of Medicine, Mansoura University, Egypt
| |
Collapse
|
32
|
Mosteo L, Storer J, Batta K, Searle EJ, Duarte D, Wiseman DH. The Dynamic Interface Between the Bone Marrow Vascular Niche and Hematopoietic Stem Cells in Myeloid Malignancy. Front Cell Dev Biol 2021; 9:635189. [PMID: 33777944 PMCID: PMC7991089 DOI: 10.3389/fcell.2021.635189] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/10/2021] [Indexed: 12/19/2022] Open
Abstract
Hematopoietic stem cells interact with bone marrow niches, including highly specialized blood vessels. Recent studies have revealed the phenotypic and functional heterogeneity of bone marrow endothelial cells. This has facilitated the analysis of the vascular microenvironment in steady state and malignant hematopoiesis. In this review, we provide an overview of the bone marrow microenvironment, focusing on refined analyses of the marrow vascular compartment performed in mouse studies. We also discuss the emerging role of the vascular niche in “inflamm-aging” and clonal hematopoiesis, and how the endothelial microenvironment influences, supports and interacts with hematopoietic cells in acute myeloid leukemia and myelodysplastic syndromes, as exemplar states of malignant myelopoiesis. Finally, we provide an overview of strategies for modulating these bidirectional interactions to therapeutic effect in myeloid malignancies.
Collapse
Affiliation(s)
- Laura Mosteo
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Joanna Storer
- Epigenetics of Haematopoiesis Group, Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom
| | - Kiran Batta
- Epigenetics of Haematopoiesis Group, Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom
| | - Emma J Searle
- Epigenetics of Haematopoiesis Group, Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom.,Department of Haematology, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Delfim Duarte
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal.,Department of Biomedicine, Faculdade de Medicina da Universidade do Porto (FMUP), Porto, Portugal.,Department of Onco-Hematology, Instituto Português de Oncologia (IPO)-Porto, Porto, Portugal
| | - Daniel H Wiseman
- Epigenetics of Haematopoiesis Group, Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom.,Department of Haematology, The Christie NHS Foundation Trust, Manchester, United Kingdom
| |
Collapse
|
33
|
López-Gil JC, Martin-Hijano L, Hermann PC, Sainz B. The CXCL12 Crossroads in Cancer Stem Cells and Their Niche. Cancers (Basel) 2021; 13:cancers13030469. [PMID: 33530455 PMCID: PMC7866198 DOI: 10.3390/cancers13030469] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/17/2021] [Accepted: 01/19/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary CXCL12 and its receptors have been extensively studied in cancer, including their influence on cancer stem cells (CSCs) and their niche. This intensive research has led to a better understanding of the crosstalk between CXCL12 and CSCs, which has aided in designing several drugs that are currently being tested in clinical trials. However, a comprehensive review has not been published to date. The aim of this review is to provide an overview on how CXCL12 axes are involved in the regulation and maintenance of CSCs, their presence and influence at different cellular levels within the CSC niche, and the current state-of-the-art of therapeutic approaches aimed to target the CXCL12 crossroads. Abstract Cancer stem cells (CSCs) are defined as a subpopulation of “stem”-like cells within the tumor with unique characteristics that allow them to maintain tumor growth, escape standard anti-tumor therapies and drive subsequent repopulation of the tumor. This is the result of their intrinsic “stem”-like features and the strong driving influence of the CSC niche, a subcompartment within the tumor microenvironment that includes a diverse group of cells focused on maintaining and supporting the CSC. CXCL12 is a chemokine that plays a crucial role in hematopoietic stem cell support and has been extensively reported to be involved in several cancer-related processes. In this review, we will provide the latest evidence about the interactions between CSC niche-derived CXCL12 and its receptors—CXCR4 and CXCR7—present on CSC populations across different tumor entities. The interactions facilitated by CXCL12/CXCR4/CXCR7 axes seem to be strongly linked to CSC “stem”-like features, tumor progression, and metastasis promotion. Altogether, this suggests a role for CXCL12 and its receptors in the maintenance of CSCs and the components of their niche. Moreover, we will also provide an update of the therapeutic options being currently tested to disrupt the CXCL12 axes in order to target, directly or indirectly, the CSC subpopulation.
Collapse
Affiliation(s)
- Juan Carlos López-Gil
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas “Alberto Sols” (IIBM), CSIC-UAM, 28029 Madrid, Spain; (J.C.L.-G.); (L.M.-H.)
- Department of Biochemistry, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain
- Chronic Diseases and Cancer, Area 3-Instituto Ramon y Cajal de Investigación Sanitaria (IRYCIS), 28029 Madrid, Spain
| | - Laura Martin-Hijano
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas “Alberto Sols” (IIBM), CSIC-UAM, 28029 Madrid, Spain; (J.C.L.-G.); (L.M.-H.)
- Department of Biochemistry, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain
- Chronic Diseases and Cancer, Area 3-Instituto Ramon y Cajal de Investigación Sanitaria (IRYCIS), 28029 Madrid, Spain
| | - Patrick C. Hermann
- Department of Internal Medicine I, Ulm University, 89081 Ulm, Germany
- Correspondence: (P.C.H.); (B.S.J.)
| | - Bruno Sainz
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas “Alberto Sols” (IIBM), CSIC-UAM, 28029 Madrid, Spain; (J.C.L.-G.); (L.M.-H.)
- Department of Biochemistry, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain
- Chronic Diseases and Cancer, Area 3-Instituto Ramon y Cajal de Investigación Sanitaria (IRYCIS), 28029 Madrid, Spain
- Correspondence: (P.C.H.); (B.S.J.)
| |
Collapse
|
34
|
Chittasupho C, Aonsri C, Imaram W. Targeted dendrimers for antagonizing the migration and viability of NALM-6 lymphoblastic leukemia cells. Bioorg Chem 2021; 107:104601. [PMID: 33476870 DOI: 10.1016/j.bioorg.2020.104601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/18/2020] [Accepted: 12/24/2020] [Indexed: 01/08/2023]
Abstract
Acute lymphoblastic leukemia (ALL) or white blood cell cancer is one of the major causes that kills many children worldwide. Although various therapeutic agents are available for ALL treatment, the new drug discovery and drug delivery system are needed to improve their effectiveness, to reduce the toxicity and side-effect, and to enhance their selectivity to target cancer cells. CXCR4 is a protein expressed on the surface of various types of cancer cell including ALL. In this work, the CXCR4-targeted PAMAM dendrimer was constructed by conjugating G5 PAMAM with a CXCR4 antagonist, LFC131. The results revealed that the LFC131-conjugated G5 PAMAM selectively targeted CXCR4 expressing leukemic precursor B cells (NALM-6) and the migration of NALM-6 cells induced by SDF-1α was inhibited at non-cytotoxic concentration. Further research based on this findings may contribute to potential anti-metastatic drugs for lymphoblastic leukemia.
Collapse
Affiliation(s)
- Chuda Chittasupho
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Mueang, Chiang Mai, Thailand
| | - Chaiyawat Aonsri
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Kasetsart University, Bangkok, Thailand
| | - Witcha Imaram
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Kasetsart University, Bangkok, Thailand; Special Research Unit for Advanced Magnetic Resonance, Department of Chemistry, Faculty of Science, Kasetsart University, Bangkok, Thailand.
| |
Collapse
|
35
|
Li L, Chai Y, Wu C, Zhao L. Chemokine receptor CXCR4: An important player affecting the molecular-targeted drugs commonly used in hematological malignancies. Expert Rev Hematol 2020; 13:1387-1396. [PMID: 33170753 DOI: 10.1080/17474086.2020.1839885] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION A variety of molecular-targeted drugs have been widely used in hematological malignancies and have shown great advances. Nevertheless, as the use of drugs in clinical practice increases, the problem of relapse or of the disease being refractory to treatment is becoming apparent. This problem is closely related to the C-X-C chemokine receptor 4 (CXCR4). AREAS COVERED This review focuses mainly on the effect of CXCR4 on molecular-targeted drug resistance in hematological malignancies as well as the clinical efficacy of CXCR4 antagonists combined with molecular-targeted drugs. Relevant literatures published between 2006 and 2020 were searched using PubMed/Medline for this review. EXPERT OPINION Monoclonal antibodies and non-antibody molecular-targeted drugs provide new therapeutic approaches for B-lineage malignancies and leukemia, but the clinical activity of these drugs is affected by CXCR4. In general, high CXCR4 expression or mutation inhibits the effects of molecular-targeted drugs, but there are exceptions, and in studies of proteasome inhibitors bortezomib (Bz) in multiple myeloma (MM), low CXCR4 expression or loss of CXCR4 was associated with Bz resistance (BzR) and poor treatment outcomes. Given that CXCR4 is a critical mediator of molecular-targeted drug resistance, numerous studies have combined molecular-targeted drugs with CXCR4 antagonists, which synergistically enhance the anti-proliferative/pro-apoptotic effect of molecular-targeted drugs.
Collapse
Affiliation(s)
- Liangliang Li
- The First Clinical Medical College of Lanzhou University , Lanzhou, Gansu, China.,Department of Hematology, Lanzhou University Second Hospital , Lanzhou, Gansu, China
| | - Ye Chai
- Department of Hematology, Lanzhou University Second Hospital , Lanzhou, Gansu, China
| | - ChongYang Wu
- Department of Hematology, Lanzhou University Second Hospital , Lanzhou, Gansu, China
| | - Li Zhao
- Department of Central Laboratory, The First Hospital of Lanzhou University , Lanzhou, Gansu, China
| |
Collapse
|
36
|
Lu C, Zhu J, Chen X, Hu Y, Xie W, Yao J, Huang S. Risk Stratification in Acute Myeloid Leukemia Using CXCR Gene Signatures: A Bioinformatics Analysis. Front Oncol 2020; 10:584766. [PMID: 33381455 PMCID: PMC7769120 DOI: 10.3389/fonc.2020.584766] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 10/09/2020] [Indexed: 12/12/2022] Open
Abstract
The role of CXC chemokine receptors in tumors has been an increasingly researched focus in recent years. However, significant prognostic values of CXCR members in acute myeloid leukemia are yet to be explored profoundly. In this study, we firstly made an analysis of the relationship of CXCR family members and AML using samples from TCGA. Our results suggested that transcriptional expressions of CXCRs serve an important role in AML. CXCR transcript expressions, except CXCR1 expression, were significantly increased in AML. It displayed the expression pattern of CXCR members in different AML subtypes according to FAB classification. The correlations of CXCR transcript expression with different genotypes and karyotypes were also present. High CXCR2 expression was found to have a significantly worse prognosis compared with that of low CXCR2 expression, and CXCR2 was also found to be an independent prognostic factor. We also established a CXCR signature to identify high-risk subgroups of patients with AML. It was an independent prognostic factor and could become a powerful method to predict the survival rate of patients.
Collapse
Affiliation(s)
- Cong Lu
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Biological Targeted Therapy Key Laboratory in Hubei, Wuhan, China
| | - Jiang Zhu
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Biological Targeted Therapy Key Laboratory in Hubei, Wuhan, China
| | - Xiangjun Chen
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Biological Targeted Therapy Key Laboratory in Hubei, Wuhan, China
| | - Yanjie Hu
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Biological Targeted Therapy Key Laboratory in Hubei, Wuhan, China
| | - Wei Xie
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Biological Targeted Therapy Key Laboratory in Hubei, Wuhan, China
| | - Junxia Yao
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Biological Targeted Therapy Key Laboratory in Hubei, Wuhan, China
| | - Shiang Huang
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Biological Targeted Therapy Key Laboratory in Hubei, Wuhan, China
| |
Collapse
|
37
|
Martin M, Mayer IA, Walenkamp AME, Lapa C, Andreeff M, Bobirca A. At the Bedside: Profiling and treating patients with CXCR4-expressing cancers. J Leukoc Biol 2020; 109:953-967. [PMID: 33089889 DOI: 10.1002/jlb.5bt1219-714r] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 08/27/2020] [Accepted: 08/27/2020] [Indexed: 12/18/2022] Open
Abstract
The chemokine receptor, C-X-C chemokine receptor type 4 (CXCR4) and its ligand, C-X-C motif chemokine 12, are key mediators of hematopoietic cell trafficking. Their roles in the proliferation and metastasis of tumor cells, induction of angiogenesis, and invasive tumor growth have been recognized for over 2 decades. CXCR4 is a promising target for imaging and therapy of both hematologic and solid tumors. To date, Sanofi Genzyme's plerixafor is the only marketed CXCR4 inhibitor (i.e., Food and Drug Administration-approved in 2008 for stem cell mobilization). However, several new CXCR4 inhibitors are now being investigated as potential therapies for a variety of fluid and solid tumors. These small molecules, peptides, and Abs include balixafortide (POL6326, Polyphor), mavorixafor (X4P-001, X4 Pharmaceuticals), motixafortide (BL-8040, BioLineRx), LY2510924 (Eli Lilly), and ulocuplumab (Bristol-Myers Squibb). Early clinical evidence has been encouraging, for example, with motixafortide and balixafortide, and the CXCR4 inhibitors appear to be generally safe and well tolerated. Molecular imaging is increasingly being used for effective patient selection before, or early during CXCR4 inhibitor treatment. The use of radiolabeled theranostics that combine diagnostics and therapeutics is an additional intriguing approach. The current status and future directions for radioimaging and treating patients with CXCR4-expressing hematologic and solid malignancies are reviewed. See related review - At the Bench: Pre-Clinical Evidence for Multiple Functions of CXCR4 in Cancer. J. Leukoc. Biol. xx: xx-xx; 2020.
Collapse
Affiliation(s)
- Miguel Martin
- Oncology Department, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense de Madrid, Madrid, Spain
| | - Ingrid A Mayer
- Division of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Annemiek M E Walenkamp
- University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Constantin Lapa
- Nuclear Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Michael Andreeff
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas, Maryland Anderson Cancer Center, Houston, Texas, USA
| | | |
Collapse
|
38
|
Cancilla D, Rettig MP, DiPersio JF. Targeting CXCR4 in AML and ALL. Front Oncol 2020; 10:1672. [PMID: 33014834 PMCID: PMC7499473 DOI: 10.3389/fonc.2020.01672] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 07/29/2020] [Indexed: 12/13/2022] Open
Abstract
The interaction of acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) blasts with the bone marrow microenvironment regulates self-renewal, growth signaling, as well as chemotherapy resistance. The chemokine receptor, CXC receptor 4 (CXCR4), with its ligand chemokine ligand 12 (CXCL12), plays a key role in the survival and migration of normal and malignant stem cells to the bone marrow. High expression of CXCR4 on AML and ALL blasts has been shown to be a predictor of poor prognosis for these diseases. Several small molecule inhibitors, short peptides, antibodies, and antibody drug conjugates have been developed for the purposes of more effective targeting and killing of malignant cells expressing CXCR4. In this review we will discuss recent results and strategies in targeting CXCR4 with these agents in patients with AML or ALL.
Collapse
Affiliation(s)
| | | | - John F. DiPersio
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
39
|
Zinngrebe J, Debatin KM, Fischer-Posovszky P. Adipocytes in hematopoiesis and acute leukemia: friends, enemies, or innocent bystanders? Leukemia 2020; 34:2305-2316. [PMID: 32474572 PMCID: PMC7449871 DOI: 10.1038/s41375-020-0886-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/15/2020] [Accepted: 05/21/2020] [Indexed: 02/07/2023]
Abstract
The bone marrow is home to well-balanced normal hematopoiesis, but also the stage of leukemia's crime. Marrow adipose tissue (MAT) is a unique and versatile component of the bone marrow niche. While the importance of MAT for bone health has long been recognized, its complex role in hematopoiesis has only recently gained attention. In this review article we summarize recent conceptual advances in the field of MAT research and how these developments impact our understanding of MAT regulation of hematopoiesis. Elucidating routes of interaction and regulation between MAT and cells of the hematopoietic system are essential to pinpoint vulnerable processes resulting in malignant transformation. The concept of white adipose tissue contributing to cancer development and progression on the cellular, metabolic, and systemic level is generally accepted. The role of MAT in malignant hematopoiesis, however, is controversial. MAT is very sensitive to changes in the patient's metabolic status hampering a clear definition of its role in different clinical situations. Here, we discuss future directions for leukemia research in the context of metabolism-induced modifications of MAT and other adipose tissues and how this might impact on leukemia cell survival, proliferation, and antileukemic therapy.
Collapse
Affiliation(s)
- Julia Zinngrebe
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Centre, D-89075, Ulm, Germany
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Centre, D-89075, Ulm, Germany
| | - Pamela Fischer-Posovszky
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Centre, D-89075, Ulm, Germany.
| |
Collapse
|
40
|
Maganti H, Visram A, Shorr R, Fulcher J, Sabloff M, Allan DS. Plerixafor in combination with chemotherapy and/or hematopoietic cell transplantation to treat acute leukemia: A systematic review and metanalysis of preclinical and clinical studies. Leuk Res 2020; 97:106442. [PMID: 32877869 DOI: 10.1016/j.leukres.2020.106442] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/21/2020] [Accepted: 08/24/2020] [Indexed: 01/01/2023]
Abstract
Leukemia-initiating cells localize to bone marrow niches via cell surface CXCR4 binding to stromal-derived factor 1 (SDF-1). Plerixafor, a CXCR4 antagonist, can mobilize and sensitize leukemia cells to cytotoxic therapy, and/or enhance the engraftment of healthy donor stem cells in the context of hematopoietic cell transplantation (HCT). A systematic review of preclinical and clinical studies was performed (updated May 1, 2020) to inform the design of definitive clinical trials and identified 19 studies. Pooled data from 10 preclinical in-vivo studies of AML and ALL in mouse models of leukemia revealed significant mobilization of leukemia cells into the peripheral circulation, decreased total blast burden and increased survival with plerixafor in addition to cytotoxic treatment compared to control animals. Two of 9 clinical studies compared outcomes to a control group. Plerixafor appears well tolerated and safe and can mobilize leukemia cells into the peripheral circulation. In patients with AML undergoing HCT, plerixafor given with the conditioning regimen appears safe and well tolerated. Engraftment, relapse and survival were not different from controls after limited follow-up. Studies in high risk patients with AML with longer follow-up are needed to understand the influence on relapse following treatment and on donor cell engraftment following HCT.
Collapse
Affiliation(s)
- Harinad Maganti
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Canada
| | - Alissa Visram
- Department of Medicine (Hematology), Faculty of Medicine, University of Ottawa, Canada
| | - Risa Shorr
- Medical Library, The Ottawa Hospital, Ottawa, Canada
| | - Jill Fulcher
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Canada; Department of Medicine (Hematology), Faculty of Medicine, University of Ottawa, Canada
| | - Mitchell Sabloff
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Canada; Department of Medicine (Hematology), Faculty of Medicine, University of Ottawa, Canada
| | - David S Allan
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Canada; Department of Medicine (Hematology), Faculty of Medicine, University of Ottawa, Canada.
| |
Collapse
|
41
|
Haltalli MLR, Lo Celso C. Targeting adhesion to the vascular niche to improve therapy for acute myeloid leukemia. Nat Commun 2020; 11:3691. [PMID: 32703951 PMCID: PMC7378234 DOI: 10.1038/s41467-020-17594-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 07/09/2020] [Indexed: 11/08/2022] Open
Abstract
Niche hijack by malignant cells is considered to be a prominent cause of disease relapse. Barbier and colleagues uncover (E)-selectin as a novel mediator of malignant cell survival and regeneration which, upon blockade, has the potential to significantly improve therapeutic outcomes.
Collapse
Affiliation(s)
- Myriam L R Haltalli
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, SW7 2AZ, London, UK
- The Francis Crick Institute, WC2A 3LY, London, UK
- Wellcome - Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, CB2 0AW, Cambridge, UK
| | - Cristina Lo Celso
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, SW7 2AZ, London, UK.
- The Francis Crick Institute, WC2A 3LY, London, UK.
| |
Collapse
|
42
|
Wang S, Wang X, Liu S, Zhang S, Wei X, Song Y, Yin Q. The CXCR4 Antagonist, AMD3100, Reverses Mesenchymal Stem Cell-Mediated Drug Resistance in Relapsed/Refractory Acute Lymphoblastic Leukemia. Onco Targets Ther 2020; 13:6583-6591. [PMID: 32753894 PMCID: PMC7352451 DOI: 10.2147/ott.s249425] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 06/07/2020] [Indexed: 12/15/2022] Open
Abstract
Purpose To investigate the role of the CXCR4/CXCL12 axis in chemotherapy resistance in refractory/relapsed (R/R) ALL patients. Methods CXCR4 expression on ALL cells from newly diagnosed or R/R ALL patients were detected using flow cytometry. The CXCR4/CXCL12 signaling pathway was blocked by the CXCR4 inhibitor AMD3100 in a co-culture model of primary drug-resistant ALL cells and umbilical cord mesenchymal stem cells (UCMSCs). Surface CXCR4 expression, apoptosis rate, and apoptosis-related protein expression in primary ALL cells under various treatments were detected. Results Of the 37 ALL patients examined, CXCR4 expression was higher in R/R patients than that in those with newly diagnosed disease. Similarly, in in vitro co-cultures of drug-resistant ALL cells with UCMSCs, the expression of CXCR4 was increased in the presence of vincristine (VCR), but reduced when VCR was combined with the CXCR4 antagonist AMD3100. Additionally, the supernatants of ALL-UCMSC co-cultures contained high CXCL12 concentrations, which were upregulated by VCR and significantly decreased by the combination of VCR plus AMD3100. Furthermore, the apoptosis rate of ALL cells significantly decreased, Bax expression was downregulated, and Bcl-2 was upregulated when ALL was co-cultured with UCMSCs compared with ALL cells alone. With the addition of VCR, the apoptosis rate mildly increased, Bax was upregulated, and Bcl-2 was downregulated. Nevertheless, the above results were further intensified, particularly Bax expression, when VCR was combined with AMD3100. Conclusion The CXCR4 antagonist could effectively reverse MSC-mediated drug resistance by blocking the CXCR4/CXCL12 axis and sensitizing leukemic cells from R/R ALL patients to chemotherapy drugs.
Collapse
Affiliation(s)
- Shan Wang
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou, People's Republic of China
| | - Xiaojiao Wang
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou, People's Republic of China
| | - Sha Liu
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou, People's Republic of China
| | - Shengnan Zhang
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou, People's Republic of China
| | - Xudong Wei
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou, People's Republic of China
| | - Yongping Song
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou, People's Republic of China
| | - Qingsong Yin
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou, People's Republic of China
| |
Collapse
|
43
|
Extracellular Adenine Nucleotides and Adenosine Modulate the Growth and Survival of THP-1 Leukemia Cells. Int J Mol Sci 2020; 21:ijms21124425. [PMID: 32580317 PMCID: PMC7352165 DOI: 10.3390/ijms21124425] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/19/2020] [Accepted: 06/20/2020] [Indexed: 12/26/2022] Open
Abstract
A new approach to improve the effectiveness of acute myeloid leukemia (AML) treatment is to use the properties of purinergic signaling molecules secreted into the bone marrow milieu in response to leukemic cell growth. Therefore, our study aimed to evaluate the effects of extracellular adenine nucleotides and adenosine on the growth and death parameters in the leukemic THP-1 cell line. Cells were exposed to ATP, ADP, AMP, adenosine and nonhydrolyzable analogues of ATP and ADP (ATPγS and ADPβS) in a 1–1000 μM broad concentration range. The basal mRNA expression of the P1 and P2 receptors was evaluated by real-time PCR. Changes in the processes of cell growth and death were assessed by flow cytometry analysis of proliferation, cell cycle and apoptosis. Chemotaxis toward stromal cell-derived factor-1 (SDF-1) was performed using the modified Boyden chamber assay, and chemokine receptor type 4 (CXCR4) surface expression was quantified by flow cytometry. We indicated several antileukemic actions. High micromolar concentrations (100–1000 μM) of extracellular adenine nucleotides and adenosine inhibit the growth of cells by arresting the cell cycle and/or inducing apoptosis. ATP is characterized by the highest potency and widest range of effects, and is responsible for the cell cycle arrest and the apoptosis induction. Compared to ATP, the effect of ADP is slightly weaker. Adenosine mostly has a cytotoxic effect, with the induction of apoptosis. The last studied nucleotide, AMP, demonstrated only a weak cytotoxic effect without affecting the cell cycle. In addition, cell migration towards SDF-1 was inhibited by low micromolar concentrations (10 μM). One of the reasons for this action of ATPγS and adenosine was a reduction in CXCR4 surface expression, but this only partially explains the mechanism of antimigratory action. In summary, extracellular adenine nucleotides and adenosine inhibit THP-1 cell growth, cause death of cells and modulate the functioning of the SDF-1/CXCR4 axis. Thus, they negatively affect the processes that are responsible for the progression of AML and the difficulties in AML treatment.
Collapse
|
44
|
Valli D, Gruszka AM, Alcalay M. Has Drug Repurposing Fulfilled its Promise in Acute Myeloid Leukaemia? J Clin Med 2020; 9:E1892. [PMID: 32560371 PMCID: PMC7356362 DOI: 10.3390/jcm9061892] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 12/16/2022] Open
Abstract
Drug repurposing is a method of drug discovery that consists of finding a new therapeutic context for an old drug. Compound identification arises from screening of large libraries of active compounds, through interrogating databases of cell line gene expression response upon treatment or by merging several types of information concerning disease-drug relationships. Although, there is a general consensus on the potential and advantages of this drug discovery modality, at the practical level to-date no non-anti-cancer repurposed compounds have been introduced into standard acute myeloid leukaemia (AML) management, albeit that preclinical validation yielded several candidates. The review presents the state-of-the-art drug repurposing approach in AML and poses the question of what has to be done in order to take a full advantage of it, both at the stage of screening design and later when progressing from the preclinical to the clinical phases of drug development. We argue that improvements are needed to model and read-out systems as well as to screening technologies, but also to more funding and trust in drug repurposing strategies.
Collapse
Affiliation(s)
- Debora Valli
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Via Adamello 16, 20 139 Milan, Italy; (D.V.); (M.A.)
| | - Alicja M. Gruszka
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Via Adamello 16, 20 139 Milan, Italy; (D.V.); (M.A.)
| | - Myriam Alcalay
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Via Adamello 16, 20 139 Milan, Italy; (D.V.); (M.A.)
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20 122 Milan, Italy
| |
Collapse
|
45
|
Méndez-Ferrer S, Bonnet D, Steensma DP, Hasserjian RP, Ghobrial IM, Gribben JG, Andreeff M, Krause DS. Bone marrow niches in haematological malignancies. Nat Rev Cancer 2020; 20:285-298. [PMID: 32112045 PMCID: PMC9912977 DOI: 10.1038/s41568-020-0245-2] [Citation(s) in RCA: 281] [Impact Index Per Article: 56.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/03/2020] [Indexed: 02/06/2023]
Abstract
Haematological malignancies were previously thought to be driven solely by genetic or epigenetic lesions within haematopoietic cells. However, the niches that maintain and regulate daily production of blood and immune cells are now increasingly being recognized as having an important role in the pathogenesis and chemoresistance of haematological malignancies. Within haematopoietic cells, the accumulation of a small number of recurrent mutations initiates malignancy. Concomitantly, specific alterations of the niches, which support haematopoietic stem cells and their progeny, can act as predisposition events, facilitating mutant haematopoietic cell survival and expansion as well as contributing to malignancy progression and providing protection of malignant cells from chemotherapy, ultimately leading to relapse. In this Perspective, we summarize our current understanding of the composition and function of the specialized haematopoietic niches of the bone marrow during health and disease. We discuss disease mechanisms (rather than malignancy subtypes) to provide a comprehensive description of key niche-associated pathways that are shared across multiple haematological malignancies. These mechanisms include primary driver mutations in bone marrow niche cells, changes associated with increased hypoxia, angiogenesis and inflammation as well as metabolic reprogramming by stromal niche cells. Consequently, remodelling of bone marrow niches can facilitate immune evasion and activation of survival pathways favouring malignant haematopoietic cell maintenance, defence against excessive reactive oxygen species and protection from chemotherapy. Lastly, we suggest guidelines for the handling and biobanking of patient samples and analysis of the niche to ensure that basic research identifying therapeutic targets can be more efficiently translated to the clinic. The hope is that integrating knowledge of how bone marrow niches contribute to haematological disease predisposition, initiation, progression and response to therapy into future clinical practice will likely improve the treatment of these disorders.
Collapse
Affiliation(s)
- Simón Méndez-Ferrer
- Wellcome Trust - Medical Research Council Cambridge Stem Cell Institute, Cambridge, UK.
- National Health Service Blood and Transplant, Cambridge, UK.
- Department of Haematology, University of Cambridge, Cambridge, UK.
| | - Dominique Bonnet
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, UK
| | - David P Steensma
- Harvard Medical School, Boston, MA, USA
- The Center for Prevention of Progression of Blood Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Robert P Hasserjian
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Irene M Ghobrial
- Harvard Medical School, Boston, MA, USA
- The Center for Prevention of Progression of Blood Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - John G Gribben
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Michael Andreeff
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Daniela S Krause
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Medicine, Frankfurt, Germany
- Goethe University Frankfurt, Frankfurt, Germany
| |
Collapse
|
46
|
Fu Z, Xiang J. Aptamers, the Nucleic Acid Antibodies, in Cancer Therapy. Int J Mol Sci 2020; 21:ijms21082793. [PMID: 32316469 PMCID: PMC7215806 DOI: 10.3390/ijms21082793] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 04/09/2020] [Accepted: 04/15/2020] [Indexed: 02/06/2023] Open
Abstract
The arrival of the monoclonal antibody (mAb) technology in the 1970s brought with it the hope of conquering cancers to the medical community. However, mAbs, on the whole, did not achieve the expected wonder in cancer therapy although they do have demonstrated successfulness in the treatment of a few types of cancers. In 1990, another technology of making biomolecules capable of specific binding appeared. This technique, systematic evolution of ligands by exponential enrichment (SELEX), can make aptamers, single-stranded DNAs or RNAs that bind targets with high specificity and affinity. Aptamers have some advantages over mAbs in therapeutic uses particularly because they have little or no immunogenicity, which means the feasibility of repeated use and fewer side effects. In this review, the general properties of the aptamer, the advantages and limitations of aptamers, the principle and procedure of aptamer production with SELEX, particularly the undergoing studies in aptamers for cancer therapy, and selected anticancer aptamers that have entered clinical trials or are under active investigations are summarized.
Collapse
Affiliation(s)
- Zhaoying Fu
- Department of Biochemistry and Molecular Biology, College of Medicine, Yanan University, Yanan 716000, China
- Correspondence: (Z.F.); (J.X.)
| | - Jim Xiang
- Division of Oncology, University of Saskatchewan, Saskatoon, SA S7N 4H4, Canada
- Correspondence: (Z.F.); (J.X.)
| |
Collapse
|
47
|
An Auristatin nanoconjugate targeting CXCR4+ leukemic cells blocks acute myeloid leukemia dissemination. J Hematol Oncol 2020; 13:36. [PMID: 32295630 PMCID: PMC7160905 DOI: 10.1186/s13045-020-00863-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 03/24/2020] [Indexed: 12/13/2022] Open
Abstract
Background Current acute myeloid leukemia (AML) therapy fails to eliminate quiescent leukemic blasts in the bone marrow, leading to about 50% of patient relapse by increasing AML burden in the bone marrow, blood, and extramedullar sites. We developed a protein-based nanoparticle conjugated to the potent antimitotic agent Auristatin E that selectively targets AML blasts because of their CXCR4 receptor overexpression (CXCR4+) as compared to normal cells. The therapeutic rationale is based on the involvement of CXCR4 overexpression in leukemic blast homing and quiescence in the bone marrow, and the association of these leukemic stem cells with minimal residual disease, dissemination, chemotherapy resistance, and lower patient survival. Methods Monomethyl Auristatin E (MMAE) was conjugated with the CXCR4 targeted protein nanoparticle T22-GFP-H6 produced in E. coli. Nanoconjugate internalization and in vitro cell viability assays were performed in CXCR4+ AML cell lines to analyze the specific antineoplastic activity through the CXCR4 receptor. In addition, a disseminated AML animal model was used to evaluate the anticancer effect of T22-GFP-H6-Auristatin in immunosuppressed NSG mice (n = 10/group). U of Mann-Whitney test was used to consider if differences were significant between groups. Results T22-GFP-H6-Auristatin was capable to internalize and exert antineoplastic effects through the CXCR4 receptor in THP-1 and SKM-1 CXCR4+ AML cell lines. In addition, repeated administration of the T22-GFP-H6-Auristatin nanoconjugate (9 doses daily) achieves a potent antineoplastic activity by internalizing specifically in the leukemic cells (luminescent THP-1) to selectively eliminate them. This leads to reduced involvement of leukemic cells in the bone marrow, peripheral blood, liver, and spleen, while avoiding toxicity in normal tissues in a luminescent disseminated AML mouse model. Conclusions A novel nanoconjugate for targeted drug delivery of Auristatin reduces significantly the acute myeloid leukemic cell burden in the bone marrow and blood and blocks its dissemination to extramedullar organs in a CXCR4+ AML model. This selective drug delivery approach validates CXCR4+ AML cells as a target for clinical therapy, not only promising to improve the control of leukemic dissemination but also dramatically reducing the severe toxicity of classical AML therapy.
Collapse
|
48
|
Izzo F, Lee SC, Poran A, Chaligne R, Gaiti F, Gross B, Murali RR, Deochand SD, Ang C, Jones PW, Nam AS, Kim KT, Kothen-Hill S, Schulman RC, Ki M, Lhoumaud P, Skok JA, Viny AD, Levine RL, Kenigsberg E, Abdel-Wahab O, Landau DA. DNA methylation disruption reshapes the hematopoietic differentiation landscape. Nat Genet 2020; 52:378-387. [PMID: 32203468 PMCID: PMC7216752 DOI: 10.1038/s41588-020-0595-4] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 02/19/2020] [Indexed: 02/07/2023]
Abstract
Mutations in genes involved in DNA methylation (DNAme; for example, TET2 and DNMT3A) are frequently observed in hematological malignancies1-3 and clonal hematopoiesis4,5. Applying single-cell sequencing to murine hematopoietic stem and progenitor cells, we observed that these mutations disrupt hematopoietic differentiation, causing opposite shifts in the frequencies of erythroid versus myelomonocytic progenitors following Tet2 or Dnmt3a loss. Notably, these shifts trace back to transcriptional priming skews in uncommitted hematopoietic stem cells. To reconcile genome-wide DNAme changes with specific erythroid versus myelomonocytic skews, we provide evidence in support of differential sensitivity of transcription factors due to biases in CpG enrichment in their binding motif. Single-cell transcriptomes with targeted genotyping showed similar skews in transcriptional priming of DNMT3A-mutated human clonal hematopoiesis bone marrow progenitors. These data show that DNAme shapes the topography of hematopoietic differentiation, and support a model in which genome-wide methylation changes are transduced to differentiation skews through biases in CpG enrichment of the transcription factor binding motif.
Collapse
Affiliation(s)
- Franco Izzo
- New York Genome Center, New York, NY, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Stanley C Lee
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Asaf Poran
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Ronan Chaligne
- New York Genome Center, New York, NY, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Federico Gaiti
- New York Genome Center, New York, NY, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Baptiste Gross
- New York Genome Center, New York, NY, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Rekha R Murali
- New York Genome Center, New York, NY, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Sunil D Deochand
- New York Genome Center, New York, NY, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Chelston Ang
- New York Genome Center, New York, NY, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Philippa Wyndham Jones
- New York Genome Center, New York, NY, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Anna S Nam
- New York Genome Center, New York, NY, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Kyu-Tae Kim
- New York Genome Center, New York, NY, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Steven Kothen-Hill
- New York Genome Center, New York, NY, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Rafael C Schulman
- New York Genome Center, New York, NY, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Michelle Ki
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Jane A Skok
- New York University Langone Health, New York, NY, USA
| | - Aaron D Viny
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ross L Levine
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ephraim Kenigsberg
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Omar Abdel-Wahab
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dan A Landau
- New York Genome Center, New York, NY, USA.
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA.
- Institute of Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
49
|
Ladikou EE, Chevassut T, Pepper CJ, Pepper AG. Dissecting the role of the CXCL12/CXCR4 axis in acute myeloid leukaemia. Br J Haematol 2020; 189:815-825. [PMID: 32135579 DOI: 10.1111/bjh.16456] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 12/03/2019] [Indexed: 02/06/2023]
Abstract
Acute myeloid leukaemia (AML) is the most common adult acute leukaemia with the lowest survival rate. It is characterised by a build-up of immature myeloid cells anchored in the protective niche of the bone marrow (BM) microenvironment. The CXCL12/CXCR4 axis is central to the pathogenesis of AML as it has fundamental control over AML cell adhesion into the protective BM niche, adaptation to the hypoxic environment, cellular migration and survival. High levels of CXCR4 expression are associated with poor relapse-free and overall survival. The CXCR4 ligand, CXCL12 (SDF-1), is expressed by multiple cells types in the BM, facilitating the adhesion and survival of the malignant clone. Blocking the CXCL12/CXCR4 axis is an attractive therapeutic strategy providing a 'multi-hit' therapy that both prevents essential survival signals and releases the AML cells from the BM into the circulation. Once out of the protective niche of the BM they would be more susceptible to destruction by conventional chemotherapeutic drugs. In this review, we disentangle the diverse roles of the CXCL12/CXCR4 axis in AML. We then describe multiple CXCR4 inhibitors, including small molecules, peptides, or monoclonal antibodies, which have been developed to date and their progress in pre-clinical and clinical trials. Finally, the review leads us to the conclusion that there is a need for further investigation into the development of a 'multi-hit' therapy that targets several signalling pathways related to AML cell adhesion and maintenance in the BM.
Collapse
Affiliation(s)
- Eleni E Ladikou
- Brighton and Sussex Medical School, University of Sussex, Brighton, UK.,Royal Sussex County Hospital, Brighton, UK
| | - Timothy Chevassut
- Brighton and Sussex Medical School, University of Sussex, Brighton, UK.,Royal Sussex County Hospital, Brighton, UK
| | - Chris J Pepper
- Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| | - Andrea Gs Pepper
- Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| |
Collapse
|
50
|
Mousavi A. CXCL12/CXCR4 signal transduction in diseases and its molecular approaches in targeted-therapy. Immunol Lett 2019; 217:91-115. [PMID: 31747563 DOI: 10.1016/j.imlet.2019.11.007] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 11/01/2019] [Accepted: 11/15/2019] [Indexed: 02/08/2023]
Abstract
Chemokines are small molecules called "chemotactic cytokines" and regulate many processes like leukocyte trafficking, homing of immune cells, maturation, cytoskeletal rearrangement, physiology, migration during development, and host immune responses. These proteins bind to their corresponding 7-membrane G-protein-coupled receptors. Chemokines and their receptors are anti-inflammatory factors in autoimmune conditions, so consider as potential targets for neutralization in such diseases. They also express by cancer cells and function as angiogenic factors, and/or survival/growth factors that enhance tumor angiogenesis and development. Among chemokines, the CXCL12/CXCR4 axis has significantly been studied in numerous cancers and autoimmune diseases. CXCL12 is a homeostatic chemokine, which is acts as an anti-inflammatory chemokine during autoimmune inflammatory responses. In cancer cells, CXCL12 acts as an angiogenic, proliferative agent and regulates tumor cell apoptosis as well. CXCR4 has a role in leukocyte chemotaxis in inflammatory situations in numerous autoimmune diseases, as well as the high levels of CXCR4, observed in different types of human cancers. These findings suggest CXCL12/CXCR4 as a potential therapeutic target for therapy of autoimmune diseases and open a new approach to targeted-therapy of cancers by neutralizing CXCL12 and CXCR4. In this paper, we reviewed the current understanding of the role of the CXCL12/CXCR4 axis in disease pathology and cancer biology, and discuss its therapeutic implications in cancer and diseases.
Collapse
|