1
|
Barton BE, Collins MK, Chau CH, Choo-Wosoba H, Venzon DJ, Steinebach C, Garchitorena KM, Shah B, Sarin EL, Gütschow M, Figg WD. Preclinical Evaluation of a Novel Series of Polyfluorinated Thalidomide Analogs in Drug-Resistant Multiple Myeloma. Biomolecules 2024; 14:725. [PMID: 38927128 PMCID: PMC11201495 DOI: 10.3390/biom14060725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/09/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
Immunomodulatory imide drugs (IMiDs) play a crucial role in the treatment landscape across various stages of multiple myeloma. Despite their evident efficacy, some patients may exhibit primary resistance to IMiD therapy, and acquired resistance commonly arises over time leading to inevitable relapse. It is critical to develop novel therapeutic options to add to the treatment arsenal to overcome IMiD resistance. We designed, synthesized, and screened a new class of polyfluorinated thalidomide analogs and investigated their anti-cancer, anti-angiogenic, and anti-inflammatory activity using in vitro and ex vivo biological assays. We identified four lead compounds that exhibit potent anti-myeloma, anti-angiogenic, anti-inflammatory properties using three-dimensional tumor spheroid models, in vitro tube formation, and ex vivo human saphenous vein angiogenesis assays, as well as the THP-1 inflammatory assay. Western blot analyses investigating the expression of proteins downstream of cereblon (CRBN) reveal that Gu1215, our primary lead candidate, exerts its activity through a CRBN-independent mechanism. Our findings demonstrate that the lead compound Gu1215 is a promising candidate for further preclinical development to overcome intrinsic and acquired IMiD resistance in multiple myeloma.
Collapse
Affiliation(s)
- Blaire E. Barton
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, Center for Cancer Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Matthew K. Collins
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, Center for Cancer Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Cindy H. Chau
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, Center for Cancer Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hyoyoung Choo-Wosoba
- Biostatics and Data Management Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - David J. Venzon
- Biostatics and Data Management Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christian Steinebach
- Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, 53121 Bonn, Germany
| | - Kathleen M. Garchitorena
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, Center for Cancer Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bhruga Shah
- Inova Heart and Vascular Institute, Inova Health System, Falls Church, VA 22042, USA
| | - Eric L. Sarin
- Inova Heart and Vascular Institute, Inova Health System, Falls Church, VA 22042, USA
| | - Michael Gütschow
- Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, 53121 Bonn, Germany
| | - William D. Figg
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, Center for Cancer Research, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
2
|
Acharya SS, Kundu CN. Havoc in harmony: Unravelling the intricacies of angiogenesis orchestrated by the tumor microenvironment. Cancer Treat Rev 2024; 127:102749. [PMID: 38714074 DOI: 10.1016/j.ctrv.2024.102749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/06/2024] [Accepted: 04/29/2024] [Indexed: 05/09/2024]
Abstract
Cancer cells merely exist in isolation; rather, they exist in an intricate microenvironment composed of blood vessels, signalling molecules, immune cells, stroma, fibroblasts, and the ECM. The TME provides a setting that is favourable for the successful growth and survivance of tumors. Angiogenesis is a multifaceted process that is essential for the growth, invasion, and metastasis of tumors. TME can be visualized as a "concert hall," where various cellular and non-cellular factors perform in a "symphony" to orchestrate tumor angiogenesis and create "Havoc" instead of "Harmony". In this review, we comprehensively summarized the involvement of TME in regulating tumor angiogenesis. Especially, we have focused on immune cells and their secreted factors, inflammatory cytokines and chemokines, and their role in altering the TME. We have also deciphered the crosstalk among various cell types that further aids the process of tumor angiogenesis. Additionally, we have highlighted the limitations of existing anti-angiogenic therapy and discussed various potential strategies that could be used to overcome these challenges and improve the efficacy of anti-angiogenic therapy.
Collapse
Affiliation(s)
- Sushree Subhadra Acharya
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University (Institute of Eminence), Campus-11, Patia, Bhubaneswar, Odisha Pin-751024, India.
| | - Chanakya Nath Kundu
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University (Institute of Eminence), Campus-11, Patia, Bhubaneswar, Odisha Pin-751024, India.
| |
Collapse
|
3
|
Hatakeyama K, Kikushige Y, Ishihara D, Yamamoto S, Kawano G, Tochigi T, Miyamoto T, Sakoda T, Christoforou A, Kunisaki Y, Fukata M, Kato K, Ito T, Handa H, Akashi K. Thrombospondin-1 is an endogenous substrate of cereblon responsible for immunomodulatory drug-induced thromboembolism. Blood Adv 2024; 8:785-796. [PMID: 38163319 PMCID: PMC10847748 DOI: 10.1182/bloodadvances.2023010080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 11/30/2023] [Accepted: 11/30/2023] [Indexed: 01/03/2024] Open
Abstract
ABSTRACT Immunomodulatory drugs (IMiDs) are key drugs for treating multiple myeloma and myelodysplastic syndrome with chromosome 5q deletion. IMiDs exert their pleiotropic effects through the interaction between cell-specific substrates and cereblon, a substrate receptor of the E3 ubiquitin ligase complex. Thus, identification of cell-specific substrates is important for understanding the effects of IMiDs. IMiDs increase the risk of thromboembolism, which sometimes results in fatal clinical outcomes. In this study, we sought to clarify the molecular mechanisms underlying IMiDs-induced thrombosis. We investigated cereblon substrates in human megakaryocytes using liquid chromatography-mass spectrometry and found that thrombospondin-1 (THBS-1), which is an inhibitor of a disintegrin-like and metalloproteinase with thrombospondin type 1 motifs 13, functions as an endogenous substrate in human megakaryocytes. IMiDs inhibited the proteasomal degradation of THBS-1 by impairing the recruitment of cereblon to THBS-1, leading to aberrant accumulation of THBS-1. We observed a significant increase in THBS-1 in peripheral blood mononuclear cells as well as larger von Willebrand factor multimers in the plasma of patients with myeloma, who were treated with IMiDs. These results collectively suggest that THBS-1 represents an endogenous substrate of cereblon. This pairing is disrupted by IMiDs, and the aberrant accumulation of THBS-1 plays an important role in the pathogenesis of IMiDs-induced thromboembolism.
Collapse
Affiliation(s)
- Kiwamu Hatakeyama
- Department of Medicine and Biosystemic Sciences, Kyushu University Graduate School of Medicine, Fukuoka, Japan
| | - Yoshikane Kikushige
- Department of Medicine and Biosystemic Sciences, Kyushu University Graduate School of Medicine, Fukuoka, Japan
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Daisuke Ishihara
- Department of Medicine and Biosystemic Sciences, Kyushu University Graduate School of Medicine, Fukuoka, Japan
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Shunsuke Yamamoto
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Gentaro Kawano
- Department of Medicine and Biosystemic Sciences, Kyushu University Graduate School of Medicine, Fukuoka, Japan
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Taro Tochigi
- Miyazaki Prefectural Miyazaki Hospital, Miyazaki, Japan
| | - Toshihiro Miyamoto
- Haematology/Respiratory Medicine, Faculty of Medicine, Institute of Medical Pharmaceutical and Health Sciences, Kanazawa University Hospital, Ishikawa, Japan
| | - Teppei Sakoda
- Department of Medicine and Biosystemic Sciences, Kyushu University Graduate School of Medicine, Fukuoka, Japan
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, Japan
| | | | - Yuya Kunisaki
- Department of Medicine and Biosystemic Sciences, Kyushu University Graduate School of Medicine, Fukuoka, Japan
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Mitsuhiro Fukata
- Department of Medicine and Biosystemic Sciences, Kyushu University Graduate School of Medicine, Fukuoka, Japan
| | - Koji Kato
- Department of Medicine and Biosystemic Sciences, Kyushu University Graduate School of Medicine, Fukuoka, Japan
| | - Takumi Ito
- Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Hiroshi Handa
- Center for Future Medical Research Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Sciences, Kyushu University Graduate School of Medicine, Fukuoka, Japan
| |
Collapse
|
4
|
Yu M, Ming H, Xia M, Fu J, Cai Z, Cui X. Identification of an angiogenesis-related risk score model for survival prediction and immunosubtype screening in multiple myeloma. Aging (Albany NY) 2024; 16:2657-2678. [PMID: 38319724 PMCID: PMC10911366 DOI: 10.18632/aging.205502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 12/27/2023] [Indexed: 02/08/2024]
Abstract
BACKGROUND Multiple myeloma (MM) is an incurable B-cell malignancy, but with the emergence of immunotherapy, a potential cure is hopeful. The individualized interaction between the tumor and bone marrow (BM) microenvironment determines the response to immunotherapy. Angiogenesis is a constant hallmark of the BM microenvironment in MM. However, little is known about the potency ability of angiogenesis-associated genes (AAGs) to regulate the immune microenvironment of MM patients. METHODS We comprehensively dissected the associations between angiogenesis and genomic landscapes, prognosis, and the immune microenvironment by integrating 36 AAGs. Immunohistochemistry was performed to verify the correlation between angiogenic factor expression and patient prognosis. Single-sample gene set enrichment analysis was applied to quantify the relative abundance of 28 infiltrating cells. The AAG score was constructed using the least absolute shrinkage and selection operator Cox regression model. RESULTS Angiogenesis was closely correlated with MM patient prognosis, and the mutation intensity of the AAGs was low. Immunohistochemistry confirmed that high microvessel density predicted poor prognosis. Three AAG clusters and two gene clusters with distinct clinical outcomes and immune characteristics were identified. The established AAG_score model performed well in predicting patient prognosis and active immunotherapy response. The high-AAG_score subgroup was characterized by reduced immune cell infiltration, poor prognosis, and inactive immunotherapy response. Multivariate analyses indicated that the AAG_score was strongly robust and independent among the prognostic variables. CONCLUSION This study revealed that angiogenesis is significantly related to MM patient prognosis and immune phenotype. Evaluating the AAG signature was conducive to predicting patient response to immunotherapy and guiding more efficacious immunotherapy strategies.
Collapse
Affiliation(s)
- Manya Yu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, China
| | - Hongquan Ming
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, China
| | - Mengting Xia
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, China
| | - Jiaqi Fu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, China
| | - Zhiguo Cai
- Department of Quality Control, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, China
| | - Xing Cui
- Department of Oncology and Hematology, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250001, China
| |
Collapse
|
5
|
Kaçmaz M, Oğuzman H. The Leucine-Rich α2-Glycoprotein-1 Levels in Patients with Multiple Myeloma. Oncol Res Treat 2023; 46:415-423. [PMID: 37527638 DOI: 10.1159/000532042] [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: 05/04/2023] [Accepted: 07/13/2023] [Indexed: 08/03/2023]
Abstract
INTRODUCTION Angiogenesis is considered important in the pathogenesis of multiple myeloma (MM), as well as in the targeted treatment of the disease. Leucine-rich α2-glycoprotein 1 (LRG1) is a protein that participates in angiogenesis and its effect on solid organ tumors has been investigated recently. This study aimed to investigate the relationship between MM and LRG1. METHODS The MM patients who admitted to Hatay Mustafa Kemal University Hematology Clinic between September 2021 and October 2022 were included in the study. The study consists of a total of 4 groups: newly diagnosed MM (NDMM), relapsed refractory MM (RRMM), MM in remission (Rem-MM), and control group. Demographic data were retrieved from hospital records. Blood samples of our study groups were centrifuged at 1,500 × g for 10 min and serum was collected. LRG1, IL-6, IL-8, TGF-β1, HIF-1α, FGF-2, and VEGF levels were analyzed in all groups by ELISA method, and statistical analysis was performed. RESULTS A total of 112 individuals, including NDMM (n: 27), RRMM (n: 18), Rem-MM (n: 42), and control group (n: 25), were enrolled in the study. Based on the analyses, the NDMM group exhibited significantly elevated levels of LRG1 (p < 0.001), TGF-1 (p < 0.001), and HIF-1α (p = 0.046, p < 0.001, and p = 0.003 compared to the RRMM, Rem-MM, and control groups, respectively) compared to the other groups. LRG1 levels were positively correlated with creatinine (r: 0.363, p = 0.001), calcium (r: 0.344, p = 0.001), total protein (r: 0.473, p < 0.001), erythrocyte sedimentation rate (r: 0.547, p < 0.001), lactate dehydrogenase (r: 0.321, p = 0.003), beta-2-microglobulin (r: 0.312, p = 0.017), IL-6 (r: 0.478, p < 0.001), IL-8 (r: 0.240, p = 0.03), TGF-β1 (r: 0.521, p < 0.001), and HIF-1α (r: 0.321, p = 0.003) levels and were negatively correlated with hemoglobin (r: -0.512, p < 0.001) and albumin (r: -0.549, p < 0.001) levels. Receiver operating characteristics (ROC) analysis revealed the association of LRG1 with the highest AUC value of 0.959 (95% CI: 0.904-1, p < 0.001) and the optimal cut-off value of 534.95 ng/mL (sensitivity: 93% and specificity: 99%) in the NDMM group compared to the control group. CONCLUSION In this study, providing data for the first time on LRG1 levels in the setting of MM. LRG1 levels were found to be significantly higher in NDMM patients and in our study discriminate this patient population from RRMM, Rem-MM, and normal controls. Therefore, LRG1 seems to a potential biomarker that should be evaluated in future studies addressing the diagnosis, staging, follow-up, prognosis, and treatment target of MM.
Collapse
Affiliation(s)
- Murat Kaçmaz
- Department of Hematology, Faculty of Medicine, Hatay Mustafa Kemal University, Antakya, Turkey
| | - Hamdi Oğuzman
- Department of Medical Biochemistry, Faculty of Medicine, Hatay Mustafa Kemal University, Antakya, Turkey
| |
Collapse
|
6
|
García-Sánchez D, González-González A, Alfonso-Fernández A, Del Dujo-Gutiérrez M, Pérez-Campo FM. Communication between bone marrow mesenchymal stem cells and multiple myeloma cells: Impact on disease progression. World J Stem Cells 2023; 15:421-437. [PMID: 37342223 PMCID: PMC10277973 DOI: 10.4252/wjsc.v15.i5.421] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 02/27/2023] [Accepted: 04/17/2023] [Indexed: 05/26/2023] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy characterized by the accumulation of immunoglobulin-secreting clonal plasma cells at the bone marrow (BM). The interaction between MM cells and the BM microenvironment, and specifically BM mesenchymal stem cells (BM-MSCs), has a key role in the pathophysiology of this disease. Multiple data support the idea that BM-MSCs not only enhance the proliferation and survival of MM cells but are also involved in the resistance of MM cells to certain drugs, aiding the progression of this hematological tumor. The relation of MM cells with the resident BM-MSCs is a two-way interaction. MM modulate the behavior of BM-MSCs altering their expression profile, proliferation rate, osteogenic potential, and expression of senescence markers. In turn, modified BM-MSCs can produce a set of cytokines that would modulate the BM microenvironment to favor disease progression. The interaction between MM cells and BM-MSCs can be mediated by the secretion of a variety of soluble factors and extracellular vesicles carrying microRNAs, long non-coding RNAs or other molecules. However, the communication between these two types of cells could also involve a direct physical interaction through adhesion molecules or tunneling nanotubes. Thus, understanding the way this communication works and developing strategies to interfere in the process, would preclude the expansion of the MM cells and might offer alternative treatments for this incurable disease.
Collapse
Affiliation(s)
- Daniel García-Sánchez
- Department of Molecular Biology_IDIVAL, Faculty of Medicine, University of Cantabria, Santander 39011, Cantabria, Spain
| | - Alberto González-González
- Department of Molecular Biology_IDIVAL, Faculty of Medicine, University of Cantabria, Santander 39011, Cantabria, Spain
| | - Ana Alfonso-Fernández
- Servicio de Traumatología y Cirugía Ortopédica, Hospital Universitario Marqués de Valdecilla, Instituto de Investigación Sanitaria Valdecilla (IDIVAL), Facultad de Medicina, Universidad de Cantabria, Santander 39008, Cantabria, Spain
| | - Mónica Del Dujo-Gutiérrez
- Department of Molecular Biology_IDIVAL, Faculty of Medicine, University of Cantabria, Santander 39011, Cantabria, Spain
| | - Flor M Pérez-Campo
- Department of Molecular Biology_IDIVAL, Faculty of Medicine, University of Cantabria, Santander 39011, Cantabria, Spain
| |
Collapse
|
7
|
Lamanuzzi A, Saltarella I, Reale A, Melaccio A, Solimando AG, Altamura C, Tamma G, Storlazzi CT, Tolomeo D, Desantis V, Mariggiò MA, Desaphy JF, Spencer A, Vacca A, Apollonio B, Frassanito MA. Uptake-Dependent and -Independent Effects of Fibroblasts-Derived Extracellular Vesicles on Bone Marrow Endothelial Cells from Patients with Multiple Myeloma: Therapeutic and Clinical Implications. Biomedicines 2023; 11:biomedicines11051400. [PMID: 37239071 DOI: 10.3390/biomedicines11051400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 04/20/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
Extracellular vesicles (EVs) have emerged as important players in cell-to-cell communication within the bone marrow (BM) of multiple myeloma (MM) patients, where they mediate several tumor-associated processes. Here, we investigate the contribution of fibroblasts-derived EVs (FBEVs) in supporting BM angiogenesis. We demonstrate that FBEVs' cargo contains several angiogenic cytokines (i.e., VEGF, HGF, and ANG-1) that promote an early over-angiogenic effect independent from EVs uptake. Interestingly, co-culture of endothelial cells from MM patients (MMECs) with FBEVs for 1 or 6 h activates the VEGF/VEGFR2, HGF/HGFR, and ANG-1/Tie2 axis, as well as the mTORC2 and Wnt/β-catenin pathways, suggesting that the early over-angiogenic effect is a cytokine-mediated process. FBEVs internalization occurs after longer exposure of MMECs to FBEVs (24 h) and induces a late over-angiogenic effect by increasing MMECs migration, chemotaxis, metalloproteases release, and capillarogenesis. FBEVs uptake activates mTORC1, MAPK, SRC, and STAT pathways that promote the release of pro-angiogenic cytokines, further supporting the pro-angiogenic milieu. Overall, our results demonstrate that FBEVs foster MM angiogenesis through dual time-related uptake-independent and uptake-dependent mechanisms that activate different intracellular pathways and transcriptional programs, providing the rationale for designing novel anti-angiogenic strategies.
Collapse
Affiliation(s)
- Aurelia Lamanuzzi
- Unit of Internal Medicine and Clinical Oncology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Ilaria Saltarella
- Unit of Internal Medicine and Clinical Oncology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, 70124 Bari, Italy
- Unit of Pharmacology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, 70121 Bari, Italy
| | - Antonia Reale
- Myeloma Research Group, Australian Centre for Blood Diseases, Central Clinical School, Monash University-Alfred Health, Melbourne, VIC 3004, Australia
| | - Assunta Melaccio
- Unit of Internal Medicine and Clinical Oncology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Antonio Giovanni Solimando
- Unit of Internal Medicine and Clinical Oncology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Concetta Altamura
- Unit of Pharmacology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, 70121 Bari, Italy
| | - Grazia Tamma
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70121 Bari, Italy
| | - Clelia Tiziana Storlazzi
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70121 Bari, Italy
| | - Doron Tolomeo
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70121 Bari, Italy
| | - Vanessa Desantis
- Unit of Pharmacology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, 70121 Bari, Italy
| | - Maria Addolorata Mariggiò
- Unit of Clinical Pathology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, 70121 Bari, Italy
| | - Jean-François Desaphy
- Unit of Pharmacology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, 70121 Bari, Italy
| | - Andrew Spencer
- Myeloma Research Group, Australian Centre for Blood Diseases, Central Clinical School, Monash University-Alfred Health, Melbourne, VIC 3004, Australia
- Malignant Haematology and Stem Cell Transplantation, Department of Haematology, Alfred Hospital, Melbourne, VIC 3004, Australia
- Department of Clinical Hematology, Monash University, Melbourne, VIC 3004, Australia
| | - Angelo Vacca
- Unit of Internal Medicine and Clinical Oncology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Benedetta Apollonio
- Unit of Internal Medicine and Clinical Oncology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Maria Antonia Frassanito
- Unit of Clinical Pathology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, 70121 Bari, Italy
| |
Collapse
|
8
|
Anloague A, Delgado-Calle J. Osteocytes: New Kids on the Block for Cancer in Bone Therapy. Cancers (Basel) 2023; 15:2645. [PMID: 37174109 PMCID: PMC10177382 DOI: 10.3390/cancers15092645] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/02/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
The tumor microenvironment plays a central role in the onset and progression of cancer in the bone. Cancer cells, either from tumors originating in the bone or from metastatic cancer cells from other body systems, are located in specialized niches where they interact with different cells of the bone marrow. These interactions transform the bone into an ideal niche for cancer cell migration, proliferation, and survival and cause an imbalance in bone homeostasis that severely affects the integrity of the skeleton. During the last decade, preclinical studies have identified new cellular mechanisms responsible for the dependency between cancer cells and bone cells. In this review, we focus on osteocytes, long-lived cells residing in the mineral matrix that have recently been identified as key players in the spread of cancer in bone. We highlight the most recent discoveries on how osteocytes support tumor growth and promote bone disease. Additionally, we discuss how the reciprocal crosstalk between osteocytes and cancer cells provides the opportunity to develop new therapeutic strategies to treat cancer in the bone.
Collapse
Affiliation(s)
- Aric Anloague
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Jesus Delgado-Calle
- Department of Physiology and Cell Biology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
9
|
Li X, Wang J, Wang Q, Luo T, Song X, Wan G, Feng Z, He X, Lei Q, Xu Y, You X, Yu L, Zhang L, Zhao L. A novel VEGFR inhibitor ZLF-095 with potent antitumor activity and low toxicity. Heliyon 2023; 9:e15152. [PMID: 37251840 PMCID: PMC10209341 DOI: 10.1016/j.heliyon.2023.e15152] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/23/2023] [Accepted: 03/28/2023] [Indexed: 05/31/2023] Open
Abstract
Angiogenesis plays a critical role in the survival, progression and metastasis of malignant tumors. Multiple factors are known to induce tumor angiogenesis, vascular endothelial growth factor (VEGF) is the most important one. Lenvatinib is an oral multi-kinase inhibitor of VEGFRs which has been approved for the treatment of various malignancies as the first-line agent by the Food and Drug Administration (FDA). It shows excellent antitumor efficacy in clinical practice. However, the adverse effects of Lenvatinib may seriously impair the therapeutic effect. Here we report the discovery and characterization of a novel VEGFR inhibitor (ZLF-095), which exhibited high activity and selectivity for VEGFR1/2/3. ZLF-095 displayed apparently antitumor effect in vitro and in vivo. We discovered that Lenvatinib could provoke fulminant ROS-caspase3-GSDME-dependent pyroptosis in GSDME-expressing cells by loss of mitochondrial membrane potential, which may be one of the reasons for Lenvatinib's toxicity. Meanwhile, ZLF-095 showed less toxicity than Lenvatinib by switching pyroptosis to apoptosis. These results suggest that ZLF-095 could become a potential angiogenesis inhibitor for cancer therapy.
Collapse
Affiliation(s)
- Xiao Li
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Jia Wang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Qianqian Wang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Tianwen Luo
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Xuejiao Song
- West China School of Public Health and West China Fourth Hospital, Sichuan University, 610000, China
| | - Guoquan Wan
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Zhanzhan Feng
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Xiaojie He
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Qian Lei
- Targeted Tracer Research and Development Laboratory, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610093, China
| | - Ying Xu
- School of Chemical Engineering, Northwest University, No.229 North Taibai Road, Xi’an, Shaanxi, 710069, China
| | - Xinyu You
- College of Chemistry and Pharmaceutical Engineering, Huanghuai University, Zhumadian 463000, China
| | - Luoting Yu
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Lidan Zhang
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, China
| | - Lifeng Zhao
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610106, China
| |
Collapse
|
10
|
Saltarella I, Altamura C, Campanale C, Laghetti P, Vacca A, Frassanito MA, Desaphy JF. Anti-Angiogenic Activity of Drugs in Multiple Myeloma. Cancers (Basel) 2023; 15:cancers15071990. [PMID: 37046651 PMCID: PMC10093708 DOI: 10.3390/cancers15071990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/20/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Angiogenesis represents a pivotal hallmark of multiple myeloma (MM) that correlates to patients’ prognosis, overall survival, and drug resistance. Hence, several anti-angiogenic drugs that directly target angiogenic cytokines (i.e., monoclonal antibodies, recombinant molecules) or their cognate receptors (i.e., tyrosine kinase inhibitors) have been developed. Additionally, many standard antimyeloma drugs currently used in clinical practice (i.e., immunomodulatory drugs, bisphosphonates, proteasome inhibitors, alkylating agents, glucocorticoids) show anti-angiogenic effects further supporting the importance of inhibiting angiogenesis from potentiating the antimyeloma activity. Here, we review the most important anti-angiogenic therapies used for the management of MM patients with a particular focus on their pharmacological profile and on their anti-angiogenic effect in vitro and in vivo. Despite the promising perspective, the direct targeting of angiogenic cytokines/receptors did not show a great efficacy in MM patients, suggesting the need to a deeper knowledge of the BM angiogenic niche for the design of novel multi-targeting anti-angiogenic therapies.
Collapse
Affiliation(s)
- Ilaria Saltarella
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Pharmacology, School of Medicine, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Concetta Altamura
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Pharmacology, School of Medicine, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Carmen Campanale
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Pharmacology, School of Medicine, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Paola Laghetti
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Pharmacology, School of Medicine, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Angelo Vacca
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, School of Medicine, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Maria Antonia Frassanito
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Clinical Pathology, School of Medicine, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Jean-François Desaphy
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Pharmacology, School of Medicine, University of Bari Aldo Moro, 70124 Bari, Italy
- Correspondence:
| |
Collapse
|
11
|
Duddu S, Bhattacharya A, Chakrabarti R, Chakravorty N, Shukla PC. Regeneration and Tissue Microenvironment. Regen Med 2023. [DOI: 10.1007/978-981-19-6008-6_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
12
|
The VEGF/VEGFR Axis Revisited: Implications for Cancer Therapy. Int J Mol Sci 2022; 23:ijms232415585. [PMID: 36555234 PMCID: PMC9779738 DOI: 10.3390/ijms232415585] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/04/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
The vascular endothelial growth factor (VEGF)/vascular endothelial growth factor receptor (VEGFR) axis is indispensable in the process of angiogenesis and has been implicated as a key driver of tumor vascularization. Consequently, several strategies that target VEGF and its cognate receptors, VEGFR-1 and VEGFR-2, have been designed to treat cancer. While therapies targeting full-length VEGF have resulted in an improvement in both overall survival and progression-free survival in various cancers, these benefits have been modest. In addition, the inhibition of VEGFRs is associated with undesirable off-target effects. Moreover, VEGF splice variants that modulate sprouting and non-sprouting angiogenesis have been identified in recent years. Cues within the tumor microenvironment determine the expression patterns of these variants. Noteworthy is that the mechanisms of action of these variants challenge the established norm of VEGF signaling. Furthermore, the aberrant expression of some of these variants has been observed in several cancers. Herein, developments in the understanding of the VEGF/VEGFR axis and the splice products of these molecules, as well as the environmental cues that regulate these variants are reviewed. Furthermore, strategies that incorporate the targeting of VEGF variants to enhance the effectiveness of antiangiogenic therapies in the clinical setting are discussed.
Collapse
|
13
|
Radnay Z, Illés Á, Udvardy M, Prohászka Z, Sinkovits G, Csányi MC, Kellermayer M, Kiss A, Hársfalvi J. Von Willebrand Factor and Platelet Levels before Conditioning Chemotherapy Indicate Bone Marrow Regeneration following Autologous Hematopoietic Stem Cell Transplantation. Transplant Cell Ther 2022; 28:830.e1-830.e7. [PMID: 36058547 DOI: 10.1016/j.jtct.2022.08.028] [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: 12/07/2021] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 12/24/2022]
Abstract
Autologous hematopoietic stem cell transplantation (HSCT) is often complicated by hemostatic and thrombotic events associated with endothelial cell injury. Thrombotic complications are affected by a disturbed balance between platelets, circulating von Willebrand factor (VWF), and its specific protease, ADAMTS13. HSCT-associated endothelial dysfunction, impaired hemostasis, and inflammation are interrelated processes, and research on the complex interplay of conditioning regimens from engraftment to bone marrow regeneration remains intensive. This prospective observational study comparing lymphoma and multiple myeloma (MM) patients who underwent autologous HSCT explored how platelet count, VWF level, ADAMTS13 activity, and C-reactive protein (CRP) level as potential markers (1) vary in response to therapy, (2) differ between the 2 groups, and (3) correlate with the remission state at 100 days after HSCT. We correlated the quantitative changes in platelet count and levels of VWF, ADAMTS13, and CRP with one another during HSCT and in the remission state in 45 patients with lymphoma and 59 patients with MM who underwent autologous HSCT between 2010 and 2013 at the University of Debrecen. Samples were collected at the start of conditioning chemotherapy, on the day of stem cell transplantation, and at 5, 11, and 100 days following HSCT. CRP levels peaked when platelet counts dropped to a minimum, and these changes were much more pronounced in the lymphoma group. VWF level was the highest, with lower ADAMTS13 activity, at platelet engraftment in both patient groups equally. Diagnostic evidence indicative of thrombotic complications was not found. In the lymphoma group, VWF level prior to conditioning had statistically significant correlations with platelet count, CRP level, and hemoglobin concentration at the time of bone marrow regeneration (P < .001) and during the remission state (P = .034). In the MM group, platelet count before conditioning was correlated with platelet count (P < .001) and white blood cell count (P = .012) at the time of bone marrow regeneration. The statistically significant correlation of the markers at the time of bone marrow regeneration with the preconditioning VWF levels in lymphoma and with the preconditioning platelet counts in MM might indicate the clinical significance of the bone marrow niches of arterioles and megakaryocytes, respectively, where the stem cells are located and regulated. Because preconditioning VWF levels are associated with remission after HSCT in lymphoma patients, VWF should be screened before conditioning, along with the markers used in HSCT protocols, to optimize personalized treatment and reduce therapeutic risks.
Collapse
Affiliation(s)
- Zita Radnay
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Árpád Illés
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Miklós Udvardy
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Prohászka
- Research Laboratory, 3rd Department of Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - György Sinkovits
- Research Laboratory, 3rd Department of Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Mária Csilla Csányi
- Department of Biophysics and Radiation Biology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Miklós Kellermayer
- Department of Biophysics and Radiation Biology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Attila Kiss
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Jolán Hársfalvi
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Hungary; Department of Biophysics and Radiation Biology, Faculty of Medicine, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
14
|
Melaccio A, Reale A, Saltarella I, Desantis V, Lamanuzzi A, Cicco S, Frassanito MA, Vacca A, Ria R. Pathways of Angiogenic and Inflammatory Cytokines in Multiple Myeloma: Role in Plasma Cell Clonal Expansion and Drug Resistance. J Clin Med 2022; 11:jcm11216491. [PMID: 36362718 PMCID: PMC9658666 DOI: 10.3390/jcm11216491] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 10/23/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022] Open
Abstract
Multiple myeloma (MM) is the second most common hematological malignancy, and despite the introduction of innovative therapies, remains an incurable disease. Identifying early and minimally or non-invasive biomarkers for predicting clinical outcomes and therapeutic responses is an active field of investigation. Malignant plasma cells (PCs) reside in the bone marrow (BM) microenvironment (BMME) which comprises cells (e.g., tumour, immune, stromal cells), components of the extracellular matrix (ECM) and vesicular and non-vesicular (soluble) molecules, all factors that support PCs’ survival and proliferation. The interaction between PCs and BM stromal cells (BMSCs), a hallmark of MM progression, is based not only on intercellular interactions but also on autocrine and paracrine circuits mediated by soluble or vesicular components. In fact, PCs and BMSCs secrete various cytokines, including angiogenic cytokines, essential for the formation of specialized niches called “osteoblastic and vascular niches”, thus supporting neovascularization and bone disease, vital processes that modulate the pathophysiological PCs–BMME interactions, and ultimately promoting disease progression. Here, we aim to discuss the roles of cytokines and growth factors in pathogenetic pathways in MM and as prognostic and predictive biomarkers. We also discuss the potential of targeted drugs that simultaneously block PCs’ proliferation and survival, PCs–BMSCs interactions and BMSCs activity, which may represent the future goal of MM therapy.
Collapse
Affiliation(s)
- Assunta Melaccio
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine “G. Baccelli”, University of Bari Medical School, 70124 Bari, Italy
- Correspondence: (A.M.); (R.R.); Tel.: +39-320-55-17-232 (A.M.)
| | - Antonia Reale
- Myeloma Research Group, Australian Centre for Blood Diseases, Central Clinical School, Monash University—Alfred Health, Melbourne 3004, Australia
| | - Ilaria Saltarella
- Department of Biomedical Sciences and Human Oncology, Pharmacology Section, University of Bari Aldo Moro Medical School, 70124 Bari, Italy
| | - Vanessa Desantis
- Department of Biomedical Sciences and Human Oncology, Pharmacology Section, University of Bari Aldo Moro Medical School, 70124 Bari, Italy
| | - Aurelia Lamanuzzi
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine “G. Baccelli”, University of Bari Medical School, 70124 Bari, Italy
| | - Sebastiano Cicco
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine “G. Baccelli”, University of Bari Medical School, 70124 Bari, Italy
| | - Maria Antonia Frassanito
- General Pathology Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70124 Bari, Italy
| | - Angelo Vacca
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine “G. Baccelli”, University of Bari Medical School, 70124 Bari, Italy
| | - Roberto Ria
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine “G. Baccelli”, University of Bari Medical School, 70124 Bari, Italy
- Correspondence: (A.M.); (R.R.); Tel.: +39-320-55-17-232 (A.M.)
| |
Collapse
|
15
|
Guo H, Yang J, Wang H, Liu X, Liu Y, Zhou K. Reshaping the tumor microenvironment: The versatility of immunomodulatory drugs in B-cell neoplasms. Front Immunol 2022; 13:1017990. [PMID: 36311747 PMCID: PMC9596992 DOI: 10.3389/fimmu.2022.1017990] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/27/2022] [Indexed: 11/24/2022] Open
Abstract
Immunomodulatory drugs (IMiDs) such as thalidomide, lenalidomide and pomalidomide are antitumor compounds that have direct tumoricidal activity and indirect effects mediated by multiple types of immune cells in the tumor microenvironment (TME). IMiDs have shown remarkable therapeutic efficacy in a set of B-cell neoplasms including multiple myeloma, B-cell lymphomas and chronic lymphocytic leukemia. More recently, the advent of immunotherapy has revolutionized the treatment of these B-cell neoplasms. However, the success of immunotherapy is restrained by immunosuppressive signals and dysfunctional immune cells in the TME. Due to the pleiotropic immunobiological properties, IMiDs have shown to generate synergetic effects in preclinical models when combined with monoclonal antibodies, immune checkpoint inhibitors or CAR-T cell therapy, some of which were successfully translated to the clinic and lead to improved responses for both first-line and relapsed/refractory settings. Mechanistically, despite cereblon (CRBN), an E3 ubiquitin ligase, is considered as considered as the major molecular target responsible for the antineoplastic activities of IMiDs, the exact mechanisms of action for IMiDs-based TME re-education remain largely unknown. This review presents an overview of IMiDs in regulation of immune cell function and their utilization in potentiating efficacy of immunotherapies across multiple types of B-cell neoplasms.
Collapse
Affiliation(s)
| | | | | | | | | | - Keshu Zhou
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
16
|
Yi Z, Ma T, Liu J, Tie W, Li Y, Bai J, Li L, Zhang L. The yin–yang effects of immunity: From monoclonal gammopathy of undetermined significance to multiple myeloma. Front Immunol 2022; 13:925266. [PMID: 35958625 PMCID: PMC9357873 DOI: 10.3389/fimmu.2022.925266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/30/2022] [Indexed: 01/10/2023] Open
Abstract
Multiple myeloma (MM) is the third most common malignant neoplasm of the hematological system. It often develops from monoclonal gammopathy of undetermined significance (MGUS) and smoldering multiple myeloma (SMM) precursor states. In this process, the immune microenvironment interacts with the MM cells to exert yin and yang effects, promoting tumor progression on the one hand and inhibiting it on the other. Despite significant therapeutic advances, MM remains incurable, and the main reason for this may be related to the complex and variable immune microenvironment. Therefore, it is crucial to investigate the dynamic relationship between the immune microenvironment and tumors, to elucidate the molecular mechanisms of different factors in the microenvironment, and to develop novel therapeutic agents targeting the immune microenvironment of MM. In this paper, we review the latest research progress and describe the dual influences of the immune microenvironment on the development and progression of MM from the perspective of immune cells and molecules.
Collapse
Affiliation(s)
- Zhigang Yi
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
- Department of Pediatric Orthopedics and Pediatrics Lanzhou University Second Hospital, Lanzhou, China
| | - Tao Ma
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
- Department of Hematology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jia Liu
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Wenting Tie
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Yanhong Li
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Jun Bai
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Lijuan Li
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
- *Correspondence: Lijuan Li, ; Liansheng Zhang,
| | - Liansheng Zhang
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
- *Correspondence: Lijuan Li, ; Liansheng Zhang,
| |
Collapse
|
17
|
Costa BA, Mouhieddine TH, Richter J. What's Old is New: The Past, Present and Future Role of Thalidomide in the Modern-Day Management of Multiple Myeloma. Target Oncol 2022; 17:383-405. [PMID: 35771402 DOI: 10.1007/s11523-022-00897-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/06/2022] [Indexed: 10/17/2022]
Abstract
Immunomodulatory drugs (IMiDs) have become an integral part of therapy for both newly diagnosed and relapsed/refractory multiple myeloma (RRMM). IMiDs bind to cereblon, leading to the degradation of proteins involved in B-cell survival and proliferation. Thalidomide, a first-generation IMiD, has little to no myelosuppressive potential, negligible renal clearance, and long-proven anti-myeloma activity. However, thalidomide's adverse effects (e.g., somnolence, constipation, and peripheral neuropathy) and the advent of more potent therapeutic options has led to the drug being less frequently used in many countries, including the US and Canada. Newer-generation IMiDs, such as lenalidomide and pomalidomide, are utilized far more frequently. In numerous previous trials, salvage therapy with thalidomide (50-200 mg/day) plus corticosteroids (with or without selected cytotoxic or targeted agents) has been shown to be effective and well-tolerated in the RRMM setting. Hence, thalidomide-based regimens remain important alternatives for heavily pretreated patients, especially for those who have no access to novel therapies and/or are not eligible for their use (due to renal failure, high-grade myelosuppression, or significant comorbidities). Ongoing and future trials may provide further insights into the current role of thalidomide, especially by comparing thalidomide-containing regimens with protocols based on newer-generation IMiDs and by investigating thalidomide's association with novel therapies (e.g., antibody-drug conjugates, bispecific antibodies, and chimeric antigen receptor T cells).
Collapse
Affiliation(s)
- Bruno Almeida Costa
- Department of Medicine, Mount Sinai Morningside and West, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tarek H Mouhieddine
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1185, New York, NY, 10029, USA
| | - Joshua Richter
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1185, New York, NY, 10029, USA.
| |
Collapse
|
18
|
Abhijit Saha, Sarker K, Ghosh A, Mishra S, Sen S. Analogue Based Design, Synthesis, Biological Evaluation, and Molecular Docking of Some Thalidomide Metabolites as Selective Cytotoxic and Antiangiogenic Agents against Multiple Myeloma. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2022. [DOI: 10.1134/s1068162022010022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
19
|
High Output Heart Failure in Multiple Myeloma: Pathogenetic Considerations. Cancers (Basel) 2022; 14:cancers14030610. [PMID: 35158878 PMCID: PMC8833382 DOI: 10.3390/cancers14030610] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/19/2022] [Accepted: 01/21/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Multiple myeloma is a plasma cell disorder that accounts for around 10% of all haematological malignancies. This neoplasia is often associated with a significant prevalence of cardiovascular complications resulting from several factors, unrelated and/or related to the disease. Among cardiovascular complications, the high output heart failure is of great importance as it is related to a worse prognosis for patients. It is important to point out that, despite the availability of more and more numerous and effective drugs, myeloma remains an incurable disease, with frequent relapses and several treatment lines, with the need, therefore, for a careful evaluation of patients, especially from a cardiological point of view. For this reason, we are proposing a comprehensive overview of different pathogenetic mechanisms responsible for high output heart failure in multiple myeloma, including artero-venous shunts, enhanced angiogenesis, glutamminolysis, hyperammonemia and hemorheological alterations, with the belief that a multidisciplinary approach, in clinical evaluation is critical for the optimal management of the patient. Abstract The high output heart failure is a clinical condition in which the systemic congestion is associated to a high output state, and it can be observed in a non-negligible percentage of hematological diseases, particularly in multiple myeloma, a condition in which the risk of adverse cardiovascular events may increase, with a worse prognosis for patients. For this reason, though an accurate literature search, we provided in this review a complete overview of different pathogenetic mechanisms responsible for high output heart failure in multiple myeloma. Indeed, this clinical finding is present in the 8% of multiple myeloma patients, and it may be caused by artero-venous shunts, enhanced angiogenesis, glutamminolysis, hyperammonemia and hemorheological alterations with increase in plasma viscosity. The high output heart failure in multiple myeloma is associated with significant morbidity and mortality, emphasizing the need for a multidisciplinary approach.
Collapse
|
20
|
Ioannidou E, Moschetta M, Shah S, Parker JS, Ozturk MA, Pappas-Gogos G, Sheriff M, Rassy E, Boussios S. Angiogenesis and Anti-Angiogenic Treatment in Prostate Cancer: Mechanisms of Action and Molecular Targets. Int J Mol Sci 2021; 22:ijms22189926. [PMID: 34576107 PMCID: PMC8472415 DOI: 10.3390/ijms22189926] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/06/2021] [Accepted: 09/13/2021] [Indexed: 12/19/2022] Open
Abstract
Prostate cancer (PC) is the most common cancer in men and the second leading cause of cancer-related death worldwide. Many therapeutic advances over the last two decades have led to an improvement in the survival of patients with metastatic PC, yet the majority of these patients still succumb to their disease. Antiagiogenic therapies have shown substantial benefits for many types of cancer but only a marginal benefit for PC. Ongoing clinical trials investigate antiangiogenic monotherapies or combination therapies. Despite the important role of angiogenesis in PC, clinical trials in refractory castration-resistant PC (CRPC) have demonstrated increased toxicity with no clinical benefit. A better understanding of the mechanism of angiogenesis may help to understand the failure of trials, possibly leading to the development of new targeted anti-angiogenic therapies in PC. These could include the identification of specific subsets of patients who might benefit from these therapeutic strategies. This paper provides a comprehensive review of the pathways involved in the angiogenesis, the chemotherapeutic agents with antiangiogenic activity, the available studies on anti-angiogenic agents and the potential mechanisms of resistance.
Collapse
Affiliation(s)
- Evangelia Ioannidou
- Department of Paediatrics and Child Health, Chelsea and Westminster Hospital, 369 Fulham Rd., London SW10 9NH, UK;
| | - Michele Moschetta
- CHUV, Lausanne University Hospital, Rue du Bugnon 21, CH-1011 Lausanne, Switzerland;
| | - Sidrah Shah
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham, Kent ME7 5NY, UK; (S.S.); (J.S.P.)
| | - Jack Steven Parker
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham, Kent ME7 5NY, UK; (S.S.); (J.S.P.)
| | - Mehmet Akif Ozturk
- Department of Medical Oncology, Sisli Memorial Hospital, Kaptan Paşa Mah. Piyale Paşa Bulv., Okmeydanı Cd. 4, Istanbul 34384, Turkey;
| | - George Pappas-Gogos
- Department of Surgery, University Hospital of Ioannina, 45111 Ioannina, Greece;
| | - Matin Sheriff
- Department of Urology, Medway NHS Foundation Trust, Windmill Road, Gillingham, Kent ME7 5NY, UK;
| | - Elie Rassy
- Department of Cancer Medicine, Gustave Roussy Institut, 94805 Villejuif, France;
| | - Stergios Boussios
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham, Kent ME7 5NY, UK; (S.S.); (J.S.P.)
- Faculty of Life Sciences & Medicine, School of Cancer & Pharmaceutical Sciences, King’s College London, London SE1 9RT, UK
- AELIA Organization, 9th Km Thessaloniki, Thermi, 57001 Thessaloniki, Greece
- Correspondence: or
| |
Collapse
|
21
|
Gynn LE, Anderson E, Robinson G, Wexler SA, Upstill-Goddard G, Cox C, May JE. Primary mesenchymal stromal cells in co-culture with leukaemic HL-60 cells are sensitised to cytarabine-induced genotoxicity, whilst leukaemic cells are protected. Mutagenesis 2021; 36:419-428. [PMID: 34505878 PMCID: PMC8633936 DOI: 10.1093/mutage/geab033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 09/09/2021] [Indexed: 12/27/2022] Open
Abstract
Tumour microenvironments are hallmarked in many cancer types. In haematological malignancies, bone marrow (BM) mesenchymal stromal cells (MSC) protect malignant cells from drug-induced cytotoxicity. However, less is known about malignant impact on supportive stroma. Notably, it is unknown whether these interactions alter long-term genotoxic damage in either direction. The nucleoside analogue cytarabine (ara-C), common in haematological therapies, remains the most effective agent for acute myeloid leukaemia, yet one third of patients develop resistance. This study aimed to evaluate the bidirectional effect of MSC and malignant cell co-culture on ara-C genotoxicity modulation. Primary MSC, isolated from patient BM aspirates for haematological investigations, and malignant haematopoietic cells (leukaemic HL-60) were co-cultured using trans-well inserts, prior to treatment with physiological dose ara-C. Co-culture genotoxic effects were assessed by micronucleus and alkaline comet assays. Patient BM cells from chemotherapy-treated patients had reduced ex vivo survival (P = 0.0049) and increased genotoxicity (P = 0.3172) than untreated patients. It was shown for the first time that HL-60 were protected by MSC from ara-C-induced genotoxicity, with reduced MN incidence in co-culture as compared to mono-culture (P = 0.0068). Comet tail intensity also significantly increased in ara-C-treated MSC with HL-60 influence (P = 0.0308). MSC sensitisation to ara-C genotoxicity was also demonstrated following co-culture with HL60 (P = 0.0116), which showed significantly greater sensitisation when MSC-HL-60 co-cultures were exposed to ara-C (P = 0.0409). This study shows for the first time that malignant HSC and MSC bidirectionally modulate genotoxicity, providing grounding for future research identifying mechanisms of altered genotoxicity in leukaemic microenvironments. MSC retain long-term genotoxic and functional damage following chemotherapy exposure. Understanding the interactions perpetuating such damage may inform modifications to reduce therapy-related complications, such as secondary malignancies and BM failure.
Collapse
Affiliation(s)
- Liana E Gynn
- Centre for Research in Biosciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK
| | - Elizabeth Anderson
- Centre for Research in Biosciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK
| | - Gareth Robinson
- Centre for Research in Biosciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK
| | - Sarah A Wexler
- Centre for Research in Biosciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK.,Royal United Hospitals Bath NHS Foundation Trust, Bath, BA1 3NG, UK
| | - Gillian Upstill-Goddard
- Centre for Research in Biosciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK.,Royal United Hospitals Bath NHS Foundation Trust, Bath, BA1 3NG, UK
| | - Christine Cox
- Centre for Research in Biosciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK.,Royal United Hospitals Bath NHS Foundation Trust, Bath, BA1 3NG, UK
| | - Jennifer E May
- Centre for Research in Biosciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK
| |
Collapse
|
22
|
Realization of Osteolysis, Angiogenesis, Immunosuppression, and Drug Resistance by Extracellular Vesicles: Roles of RNAs and Proteins in Their Cargoes and of Ectonucleotidases of the Immunosuppressive Adenosinergic Noncanonical Pathway in the Bone Marrow Niche of Multiple Myeloma. Cancers (Basel) 2021; 13:cancers13122969. [PMID: 34199285 PMCID: PMC8231946 DOI: 10.3390/cancers13122969] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 05/31/2021] [Accepted: 06/10/2021] [Indexed: 12/26/2022] Open
Abstract
Simple Summary Multiple myeloma (MM) is a disease that extensively involves bone, and angiogenesis and immunosuppression are important processes in the development of MM. Proteasome inhibitors and immunomodulatory drugs remarkably improve the survival of MM patients. However, MM is still an incurable disease that rapidly becomes resistant to these drugs. There is robust evidence that extracellular vesicles (EVs) contribute to cancer metastasis. Osteoclasts, in addition to immunosuppressive cells in the bone marrow (BM), are key players in osteolysis and immunosuppression. BM stromal cells and MM cells secrete EVs through which they communicate with each other: EVs, in fact, contain proteins, small RNAs, and long non-coding RNAs that mediate this communication and contribute to angiogenesis, osteolysis, and cancer dissemination and drug resistance. Ectoenzymes are expressed in myeloma cells, osteoclasts, and stromal cells and produce immunosuppressive adenosine. Recently, an antibody targeting CD38, an ectoenzyme, has been shown to improve the survival of patients with MM. Thus, understanding the properties of EV and ectoenzymes will help elucidate key processes of MM development. Abstract Angiogenesis and immunosuppression promote multiple myeloma (MM) development, and osteolysis is a primary feature of MM. Although immunomodulatory drugs and proteasome inhibitors (PIs) markedly improve the survival of patients with MM, this disease remains incurable. In the bone marrow niche, a chain of ectoenzymes, including CD38, produce immunosuppressive adenosine, inhibiting T cell proliferation as well as immunosuppressive cells. Therefore, anti-CD38 antibodies targeting myeloma cells have the potential to restore T cell responses to myeloma cells. Meanwhile extracellular vesicles (EVs) containing microRNAs, proteins such as cytokines and chemokines, long noncoding RNAs, and PIWI-interacting RNAs have been shown to act as communication tools in myeloma cell/microenvironment interactions. Via EVs, mesenchymal stem cells allow myeloma cell dissemination and confer PI resistance, whereas myeloma cells promote angiogenesis, myeloid-derived suppressor cell proliferation, and osteoclast differentiation and inhibit osteoblast differentiation. In this review, to understand key processes of MM development involving communication between myeloma cells and other cells in the tumor microenvironment, EV cargo and the non-canonical adenosinergic pathway are introduced, and ectoenzymes and EVs are discussed as potential druggable targets for the treatment of MM patients.
Collapse
|
23
|
Tomaipitinca L, Russo E, Bernardini G. NK cell surveillance of hematological malignancies. Therapeutic implications and regulation by chemokine receptors. Mol Aspects Med 2021; 80:100968. [PMID: 34045078 DOI: 10.1016/j.mam.2021.100968] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/06/2021] [Accepted: 05/14/2021] [Indexed: 11/26/2022]
Abstract
NK cells are circulating innate lymphoid cells that constantly move from bloodstream into tissues, exerting several functions including tumor surveillance. For this reason, NK cells are considered attractive target for cancer immunotherapy. Several strategies are employed to harness NK cell efficacy especially in hematological tumors, including adoptive transfer, genetic manipulation to overexpress chimeric antigen receptors and cytokine or immunomodulatory drug treatments of ex-vivo cultivated and expanded NK cells. Several chemokine receptors support NK cell tissue homing and are required for efficient tumor infiltration. Nevertheless, chemokine receptor expression is often insufficient, or their respective ligands may not be expressed in the tumor microenvironment, thus limiting NK cell localization at the tumor site. Therefore, strategies to implement expression or promote the function of the correct chemokine receptor/ligand axes have been employed in the last years with promising results in preclinical models. In this review, we discuss how chemokine receptors and their ligands regulate the trafficking and localization of NK cells in hematological tumors and how the chemokine function can be manipulated to improve current therapeutic approaches.
Collapse
Affiliation(s)
- Luana Tomaipitinca
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Pasteur Institute Italia-Fondazione Cenci Bolognetti, Viale Regina Elena 291, 00161, Rome, Italy
| | - Eleonora Russo
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Pasteur Institute Italia-Fondazione Cenci Bolognetti, Viale Regina Elena 291, 00161, Rome, Italy
| | - Giovanni Bernardini
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Pasteur Institute Italia-Fondazione Cenci Bolognetti, Viale Regina Elena 291, 00161, Rome, Italy.
| |
Collapse
|
24
|
Yousefi Y, Mortezania Z, Abolmolouki M, Daemi A. A novel form of anti-angiogenic molecular antibody drug induces apoptosis in myeloma cells after cultivation upon endothelial feeder cells. Leuk Res 2021; 108:106617. [PMID: 34022745 DOI: 10.1016/j.leukres.2021.106617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 11/30/2022]
Abstract
The present study aimed to investigate the ability of a novel form of the anti-angiogenic molecular antibody drug to induce Myeloma cell death after cultivation upon endothelial feeder cells. Bevacizumab-loaded chitosan (BCS) nanoparticles (NPs) were prepared by the ionic gelation method. Human U266 cell line and human umbilical vein endothelial cells were co-cultured for 72 h and treated with BCS nanoparticles (10μM) to study their impact on inhibition of cell growth and induction of apoptosis. Death assessments, P53 pro-apoptotic marker expression and the VEGF level were investigated by flow-cytometric analyses of the Annexin V, immunocytochemistry and ELISA, respectively. The endothelial monolayer co-culture showed protection of myeloma cells from apoptosis when exposed to NPs or without any treatment. In present of bevacizumab, the VEGF factor was effectively suppressed, and the p53 expression was significantly increased in bevacizumab-treated myeloma cells co-cultured with HUVECs, compared to other groups. BCS was capable of disturbing the effective interconnections of myeloma cells and HUVECs as supportive cells. Disruptions of tumor cells' connections with their microenvironment and blocking their possible supportive pathways might be a potential strategy to eradicate tumor cells and finally cure such types of cancer.
Collapse
Affiliation(s)
- Yavar Yousefi
- Iranian Center of Neurological Research, Tehran University of Medical Sciences, Tehran, Iran.
| | - Zohreh Mortezania
- Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mojdeh Abolmolouki
- Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| | - Amin Daemi
- Department of Medical Biochemistry, Faculty of Medicine, Çukurova University, Adana, Turkey.
| |
Collapse
|
25
|
Ronca R, Taranto S, Corsini M, Tobia C, Ravelli C, Rezzola S, Belleri M, De Cillis F, Cattaneo A, Presta M, Giacomini A. Pentraxin 3 Inhibits the Angiogenic Potential of Multiple Myeloma Cells. Cancers (Basel) 2021; 13:cancers13092255. [PMID: 34066669 PMCID: PMC8125855 DOI: 10.3390/cancers13092255] [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: 04/02/2021] [Revised: 04/30/2021] [Accepted: 05/05/2021] [Indexed: 11/30/2022] Open
Abstract
Simple Summary Bone marrow (BM) angiogenesis represents a key aspect in the progression of multiple myeloma (MM) and is strictly linked to the balance between pro-angiogenic and anti-angiogenic players produced by both neoplastic and stromal components. It has been shown that Fibroblast Growth Factors (FGFs) play a pivotal role in the angiogenic switch occurring during MM progression. Accordingly, the natural FGF antagonist Long Pentraxin 3 (PTX3) is able to reduce the activation of BM stromal components induced by FGFs. This work explores, for the first time, the anti-angiogenic role of PTX3 produced by MM cells demonstrating that the inducible expression of PTX3 is able to impair MM neovascularization, the onset of a proficient BM vascular niche and, ultimately, to impair tumor growth and dissemination. Abstract During multiple myeloma (MM) progression the activation of the angiogenic process represents a key step for the formation of the vascular niche, where different stromal components and neoplastic cells collaborate and foster tumor growth. Among the different pro-angiogenic players, Fibroblast Growth Factor 2 (FGF2) plays a pivotal role in BM vascularization occurring during MM progression. Long Pentraxin 3 (PTX3), a natural FGF antagonist, is able to reduce the activation of stromal components promoted by FGF2 in various in vitro models. An increased FGF/PTX3 ratio has also been found to occur during MM evolution, suggesting that restoring the “physiological” FGF/PTX3 ratio in plasma cells and BM stromal cells (BMSCs) might impact MM. In this work, taking advantage of PTX3-inducible human MM models, we show that PTX3 produced by tumor cells is able to restore a balanced FGF/PTX3 ratio sufficient to prevent the activation of the FGF/FGFR system in endothelial cells and to reduce the angiogenic capacity of MM cells in different in vivo models. As a result of this anti-angiogenic activity, PTX3 overexpression causes a significant reduction of the tumor burden in both subcutaneously grafted and systemic MM models. These data pave the way for the exploitation of PTX3-derived anti-angiogenic approaches in MM.
Collapse
Affiliation(s)
- Roberto Ronca
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (S.T.); (M.C.); (C.T.); (C.R.); (S.R.); (M.B.); (M.P.)
- Correspondence: (R.R.); (A.G.)
| | - Sara Taranto
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (S.T.); (M.C.); (C.T.); (C.R.); (S.R.); (M.B.); (M.P.)
| | - Michela Corsini
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (S.T.); (M.C.); (C.T.); (C.R.); (S.R.); (M.B.); (M.P.)
| | - Chiara Tobia
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (S.T.); (M.C.); (C.T.); (C.R.); (S.R.); (M.B.); (M.P.)
| | - Cosetta Ravelli
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (S.T.); (M.C.); (C.T.); (C.R.); (S.R.); (M.B.); (M.P.)
| | - Sara Rezzola
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (S.T.); (M.C.); (C.T.); (C.R.); (S.R.); (M.B.); (M.P.)
| | - Mirella Belleri
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (S.T.); (M.C.); (C.T.); (C.R.); (S.R.); (M.B.); (M.P.)
| | - Floriana De Cillis
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (F.D.C.); (A.C.)
| | - Annamaria Cattaneo
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (F.D.C.); (A.C.)
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20122 Milan, Italy
| | - Marco Presta
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (S.T.); (M.C.); (C.T.); (C.R.); (S.R.); (M.B.); (M.P.)
| | - Arianna Giacomini
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (S.T.); (M.C.); (C.T.); (C.R.); (S.R.); (M.B.); (M.P.)
- Correspondence: (R.R.); (A.G.)
| |
Collapse
|
26
|
Wang X, He L, Huang X, Zhang S, Cao W, Che F, Zhu Y, Dai J. Recent Progress of Exosomes in Multiple Myeloma: Pathogenesis, Diagnosis, Prognosis and Therapeutic Strategies. Cancers (Basel) 2021; 13:cancers13071635. [PMID: 33915822 PMCID: PMC8037106 DOI: 10.3390/cancers13071635] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/20/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary In the pathogenesis of multiple myeloma (MM), some exosomes act on different cells in the bone marrow microenvironment, which can create an environment conducive to the survival and growth of MM cells. In addition, due to the abnormal expression of cargos in the exosomes of MM patients, exosomes may help with the diagnosis and prognosis of MM. In contrast to traditional nanomaterials, exosomes exhibit very good safety, biocompatibility, stability and biodegradability, which shows their potential for delivering anti-cancer drugs and cancer vaccines. Given the research in recent decades, exosomes are becoming increasingly relevant to MM. Although exosomes have not been applied in the clinic for help with diagnosing, prognosticating or providing therapy for MM, they are very promising for clinical applications concerning MM, which will possibly materialize in the near future. Therefore, this review is worth reading for further understanding of the important roles of exosomes in MM.. Abstract Multiple myeloma (MM) is a hematological malignancy that is still incurable. The bone marrow microenvironment (BMM), with cellular and non-cellular components, can create a favorable environment for the survival, proliferation and migration of MM cells, which is the main reason for the failure of MM therapies. Many studies have demonstrated that exosomes play an important role in the tumor-supportive BMM. Exosomes are nanoscale vesicles that can be released by various cells. Some exosomes contribute to the pathogenesis and progression of MM. MM-derived exosomes act on different cells in the BMM, thereby creating an environment conducive to the survival and growth of MM cells. Owing to the important roles of exosomes in the BMM, targeting the secretion of exosomes may become an effective therapeutic strategy for MM. In addition, the abnormal expression of “cargos” in the exosomes of MM patients may be used to diagnose MM or used as part of a screen for the early prognoses of MM patients. Exosomes also have good biological properties, including safety, biocompatibility, stability and biodegradability. Therefore, the encapsulation of anti-cancer drugs in exosomes, along with surface modifications of exosomes with targeting molecules, are very promising strategies for cancer therapies—particularly for MM. In addition, DC-derived exosomes (DC-EXs) can express MHC-I, MHC-II and T cell costimulatory molecules. Therefore, DC-EXs may be used as a nanocarrier to deliver cancer vaccines in MM. This review summarizes the recent progress of exosome research regarding the pathogenesis of, diagnosis of, prognosis of and therapeutic strategies for MM.
Collapse
Affiliation(s)
- Xi Wang
- Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu 610000, China; (X.W.); (L.H.); (X.H.); (S.Z.); (F.C.)
| | - Lin He
- Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu 610000, China; (X.W.); (L.H.); (X.H.); (S.Z.); (F.C.)
| | - Xiaobing Huang
- Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu 610000, China; (X.W.); (L.H.); (X.H.); (S.Z.); (F.C.)
| | - Shasha Zhang
- Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu 610000, China; (X.W.); (L.H.); (X.H.); (S.Z.); (F.C.)
| | - Wanjun Cao
- School of Pharmacy, North Sichuan Medical College, Nanchong 637000, China;
| | - Feifei Che
- Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu 610000, China; (X.W.); (L.H.); (X.H.); (S.Z.); (F.C.)
| | - Yizhun Zhu
- State Key Laboratory of Quality Research in Chinese Medicine & School of Pharmacy, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau 999078, China
- Correspondence: (Y.Z.); (J.D.); Tel.: +86-15756317270 (J.D.)
| | - Jingying Dai
- Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu 610000, China; (X.W.); (L.H.); (X.H.); (S.Z.); (F.C.)
- Correspondence: (Y.Z.); (J.D.); Tel.: +86-15756317270 (J.D.)
| |
Collapse
|
27
|
Tian X, Sun M, Wu H, Chen C, Li H, Qiu S, Wang T, Han J, Xiao Q, Chen K. Exosome-derived miR-let-7c promotes angiogenesis in multiple myeloma by polarizing M2 macrophages in the bone marrow microenvironment. Leuk Res 2021; 105:106566. [PMID: 33848709 DOI: 10.1016/j.leukres.2021.106566] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/03/2021] [Accepted: 03/17/2021] [Indexed: 12/12/2022]
Abstract
Angiogenesis is an integral part of the multiple myeloma (MM) microenvironment, and affects tumorigenesis, progression, invasion, and metastasis. Exosomes are essential for cell-cell communication and help in regulating the bone marrow microenvironment. Herein, we investigated macrophage polarization and angiogenesis in MM in vitro via exosome-derived miR-let-7c. We observed that exosomal miR-let-7c secreted by mesenchymal stem cells promoted M2 macrophage polarization, thereby enhancing angiogenesis in the bone marrow microenvironment. Suppressing miR-let-7c expression significantly inhibited vascular endothelial cell function in myeloma. Thus, exosomal miR-let-7c may be a reliable biomarker for early prediction of tumor progression and a promising therapeutic target for MM.
Collapse
Affiliation(s)
- Xiangyu Tian
- School of Basic Medical Sciences, Zhengzhou University, No. 100 Ke Xue Avenue, Zhengzhou, 450000, China; Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, No. 1 Jian She Dong Avenue, Zhengzhou, 450000, China; Henan Key Laboratory of Tumor Pathology, Zhengzhou University, No. 40 Da Xue Avenue, Zhengzhou, 450000, China
| | - Miaomiao Sun
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jian She Dong Avenue, Zhengzhou, 450000, China
| | - Han Wu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, No. 1 Jian She Dong Avenue, Zhengzhou, 450000, China
| | - Chao Chen
- School of Basic Medical Sciences, Zhengzhou University, No. 100 Ke Xue Avenue, Zhengzhou, 450000, China
| | - Hui Li
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jian She Dong Avenue, Zhengzhou, 450000, China
| | - Sen Qiu
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, No. 40 Da Xue Avenue, Zhengzhou, 450000, China
| | - Tong Wang
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, No. 40 Da Xue Avenue, Zhengzhou, 450000, China
| | - Junya Han
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, No. 40 Da Xue Avenue, Zhengzhou, 450000, China
| | - Qiankun Xiao
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, No. 40 Da Xue Avenue, Zhengzhou, 450000, China
| | - Kuisheng Chen
- School of Basic Medical Sciences, Zhengzhou University, No. 100 Ke Xue Avenue, Zhengzhou, 450000, China; Henan Key Laboratory of Tumor Pathology, Zhengzhou University, No. 40 Da Xue Avenue, Zhengzhou, 450000, China; Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jian She Dong Avenue, Zhengzhou, 450000, China.
| |
Collapse
|
28
|
Parmar D, Apte M. Angiopoietin inhibitors: A review on targeting tumor angiogenesis. Eur J Pharmacol 2021; 899:174021. [PMID: 33741382 DOI: 10.1016/j.ejphar.2021.174021] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 02/24/2021] [Accepted: 03/10/2021] [Indexed: 02/08/2023]
Abstract
Angiogenesis is the process of formation of new blood vessels from existing ones. Vessels serve the purpose of providing oxygen, nutrients and removal of waste from the cells. The physiological angiogenesis is a normal process and is required in the embryonic development, wound healing, menstrual cycle. For homeostasis, balance of pro angiogenic factors and anti angiogenic factors like is important. Their imbalance causes a process known as "angiogenic switch" which leads to various pathological conditions like inflammation, tumor and restenosis. Like normal cells, tumor cells also require oxygen and nutrients to grow which is provided by tumor angiogenesis. Hence angiogenic process can be inhibited to prevent tumor growth. This gives rise to study of anti angiogenic drugs. Currently approved anti angiogenic drugs are mostly VEGF inhibitors, but VEGF inhibitors have certain limitations like toxicity, low progression free survival (PFS), and resistance to anti VEGF therapy. This article focuses on angiopoietins as alternative and potential targets for anti angiogenic therapy. Angiopoietins are ligands of Tie receptor and play a crucial role in angiogenesis, their inhibition can prevent many tumor growths even on later stages of development. We present current clinical and preclinical stages of angiopoietin inhibitors. Drugs studied in the article are selective as well as non-selective inhibitors of angiopoietin 2 like Trebananib (AMG 386), AMG 780, REGN 910, CVX 060, MEDI 3617 and dual inhibitors of angiopoietin 2 and VEGF like Vanucizumab and RG7716. The angiopoietin inhibitors show promising results alone and in combination with VEGF inhibitors in various malignancies.
Collapse
Affiliation(s)
- Digna Parmar
- Department of Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle, Maharashtra, India.
| | - Madhavi Apte
- Department of Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle, Maharashtra, India.
| |
Collapse
|
29
|
Lopes R, Ferreira BV, Caetano J, Barahona F, Carneiro EA, João C. Boosting Immunity against Multiple Myeloma. Cancers (Basel) 2021; 13:1221. [PMID: 33799565 PMCID: PMC8001641 DOI: 10.3390/cancers13061221] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/12/2021] [Accepted: 03/01/2021] [Indexed: 01/10/2023] Open
Abstract
Despite the improvement of patient's outcome obtained by the current use of immunomodulatory drugs, proteasome inhibitors or anti-CD38 monoclonal antibodies, multiple myeloma (MM) remains an incurable disease. More recently, the testing in clinical trials of novel drugs such as anti-BCMA CAR-T cells, antibody-drug conjugates or bispecific antibodies broadened the possibility of improving patients' survival. However, thus far, these treatment strategies have not been able to steadily eliminate all malignant cells, and the aim has been to induce a long-term complete response with minimal residual disease (MRD)-negative status. In this sense, approaches that target not only myeloma cells but also the surrounding microenvironment are promising strategies to achieve a sustained MRD negativity with prolonged survival. This review provides an overview of current and future strategies used for immunomodulation of MM focusing on the impact on bone marrow (BM) immunome.
Collapse
Affiliation(s)
- Raquel Lopes
- Lymphoma and Myeloma Research Programme, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal; (R.L.); (B.V.F.); (J.C.); (F.B.); (E.A.C.)
- Faculty of Medicine, University of Lisbon, 1649-028 Lisbon, Portugal
| | - Bruna Velosa Ferreira
- Lymphoma and Myeloma Research Programme, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal; (R.L.); (B.V.F.); (J.C.); (F.B.); (E.A.C.)
- Faculty of Medical Sciences, NOVA Medical School, 1169-056 Lisbon, Portugal
| | - Joana Caetano
- Lymphoma and Myeloma Research Programme, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal; (R.L.); (B.V.F.); (J.C.); (F.B.); (E.A.C.)
- Faculty of Medical Sciences, NOVA Medical School, 1169-056 Lisbon, Portugal
- Hemato-Oncology Department, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| | - Filipa Barahona
- Lymphoma and Myeloma Research Programme, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal; (R.L.); (B.V.F.); (J.C.); (F.B.); (E.A.C.)
- Faculty of Medical Sciences, NOVA Medical School, 1169-056 Lisbon, Portugal
| | - Emilie Arnault Carneiro
- Lymphoma and Myeloma Research Programme, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal; (R.L.); (B.V.F.); (J.C.); (F.B.); (E.A.C.)
| | - Cristina João
- Lymphoma and Myeloma Research Programme, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal; (R.L.); (B.V.F.); (J.C.); (F.B.); (E.A.C.)
- Faculty of Medical Sciences, NOVA Medical School, 1169-056 Lisbon, Portugal
- Hemato-Oncology Department, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| |
Collapse
|
30
|
Russignan A, Dal Collo G, Bagnato A, Tamassia N, Bugatti M, Belleri M, Lorenzi L, Borsi E, Bazzoni R, Gottardi M, Terragna C, Vermi W, Giacomini A, Presta M, Cassatella MA, Krampera M, Tecchio C. Targeting the Endothelin-1 Receptors Curtails Tumor Growth and Angiogenesis in Multiple Myeloma. Front Oncol 2021; 10:600025. [PMID: 33489901 PMCID: PMC7820698 DOI: 10.3389/fonc.2020.600025] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 11/16/2020] [Indexed: 12/17/2022] Open
Abstract
The endothelin-1 (ET-1) receptors were recently found to mediate pro-survival functions in multiple myeloma (MM) cells in response to autocrine ET-1. This study investigated the effectiveness of macitentan, a dual ET-1 receptor antagonist, in MM treatment, and the mechanisms underlying its activities. Macitentan affected significantly MM cell (RPMI-8226, U266, KMS-12-PE) survival and pro-angiogenic cytokine release by down-modulating ET-1-activated MAPK/ERK and HIF-1α pathways, respectively. HIF-1α silencing abrogated the ET-1 mediated induction of genes encoding for pro-angiogenic cytokines such as VEGF-A, IL-8, Adrenomedullin, and ET-1 itself. Upon exposure to macitentan, MM cells cultured in the presence of the hypoxia-mimetic agent CoCl2, exogenous ET-1, or CoCl2 plus ET-1, down-regulated HIF-1α and the transcription and release of downstream pro-angiogenic cytokines. Consistently, macitentan limited significantly the basal pro-angiogenic activity of RPMI-8226 cells in chorioallantoic membrane assay. In xenograft mouse models, established by injecting NOG mice either via intra-caudal vein with U266 or subcutaneously with RPMI-8226 cells, macitentan reduced effectively the number of MM cells infiltrating bone marrow, and the size and microvascular density of subcutaneous MM tumors. ET-1 receptors targeting by macitentan represents an effective anti-proliferative and anti-angiogenic therapeutic approach in preclinical settings of MM.
Collapse
Affiliation(s)
- Anna Russignan
- Section of Hematology and Bone-Marrow Transplant Unit, Department of Medicine, University of Verona, Verona, Italy
| | - Giada Dal Collo
- Section of Hematology and Bone-Marrow Transplant Unit, Department of Medicine, University of Verona, Verona, Italy
| | - Anna Bagnato
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Nicola Tamassia
- Section of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| | - Mattia Bugatti
- Section of Pathology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Mirella Belleri
- Experimental Oncology and Immunology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Luisa Lorenzi
- Section of Pathology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Enrica Borsi
- Department of Experimental Diagnostic and Specialty Medicine (DIMES), "L. and A. Seràgnoli", Bologna University, Bologna, Italy
| | - Riccardo Bazzoni
- Section of Hematology and Bone-Marrow Transplant Unit, Department of Medicine, University of Verona, Verona, Italy
| | | | - Carolina Terragna
- Department of Experimental Diagnostic and Specialty Medicine (DIMES), "L. and A. Seràgnoli", Bologna University, Bologna, Italy
| | - William Vermi
- Section of Pathology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Arianna Giacomini
- Experimental Oncology and Immunology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Marco Presta
- Experimental Oncology and Immunology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | | - Mauro Krampera
- Section of Hematology and Bone-Marrow Transplant Unit, Department of Medicine, University of Verona, Verona, Italy
| | - Cristina Tecchio
- Section of Hematology and Bone-Marrow Transplant Unit, Department of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
31
|
Iannazzo D, Ettari R, Giofrè S, Eid AH, Bitto A. Recent Advances in Nanotherapeutics for Multiple Myeloma. Cancers (Basel) 2020; 12:cancers12113144. [PMID: 33120945 PMCID: PMC7693822 DOI: 10.3390/cancers12113144] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 10/26/2020] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Nanotherapeutics are useful tools to improve the deliverability of drugs, especially anti-cancer drugs that need to target specific cells. Several approaches have been studied for multiple myeloma, considering that immune cells are not easy to target with the available drugs. These pharmacological agents are administered in various combinations using Thalidomide (or Lenalidomide, Pomalidomide), corticosteroids (Dexamethasone), proteasome inhibitors (Bortezomib, Carfilzomib, Ixazomib), deacetylase inhibitors (Panobinostat), and monoclonal antibodies (Elotuzumab, Daratumumab). As all drugs these agents might have serious side effects and in addition, the reliance on stochastic events to deliver drugs to tumors reduces their effectiveness either through rapid clearance from blood or inadequate concentration in cancer cells. To address these issues liposomes, micelles, polymeric nanoparticles, inorganic nanoparticles, and carbon-based nanomaterials have been successfully tested in vivo and can be considered as useful tools to improve delivery of active pharmaceuticals that show poor bioavailability or poor internalization into myeloma cells. Abstract Anticancer therapies cannot be included in a one-size-fits-all scenario; it is imperative to adapt therapies to the tumor molecular profile and most importantly to develop target-specific therapeutics. Nanotherapeutics can combine molecular imaging with molecular therapy in order to provide the maximum benefit to patients in terms of disease prevention, identification, and treatment. Nanotechnology applied to therapy provides numerous advantages in diagnostics and in drug delivery, especially for those malignant cells that are difficult to target or for drugs with poor bioavailability, such as those used for multiple myeloma (MM). This review summarizes the recent advances in the development of nanoparticle-based systems for the treatment of MM, taking into account the methods used for their functionalization, biocompatibility, and anticancer activity.
Collapse
Affiliation(s)
- Daniela Iannazzo
- Department of Engineering, University of Messina, 98166 Messina, Italy;
| | - Roberta Ettari
- Department of Chemical, Biological, Pharmaceutical and Environmental Chemistry, University of Messina, 98165 Messina, Italy; (R.E.); (S.G.)
| | - Salvatore Giofrè
- Department of Chemical, Biological, Pharmaceutical and Environmental Chemistry, University of Messina, 98165 Messina, Italy; (R.E.); (S.G.)
| | - Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, 2713 Doha, Qatar;
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, 2713 Doha, Qatar
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, 11-0236 Beirut, Lebanon
| | - Alessandra Bitto
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
- Correspondence:
| |
Collapse
|
32
|
Testa U, Pelosi E, Castelli G. Endothelial Progenitors in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1263:85-115. [PMID: 32588325 DOI: 10.1007/978-3-030-44518-8_7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tumor vascularization refers to the formation of new blood vessels within a tumor and is considered one of the hallmarks of cancer. Tumor vessels supply the tumor with oxygen and nutrients, required to sustain tumor growth and progression, and provide a gateway for tumor metastasis through the blood or lymphatic vasculature. Blood vessels display an angiocrine capacity of supporting the survival and proliferation of tumor cells through the production of growth factors and cytokines. Although tumor vasculature plays an essential role in sustaining tumor growth, it represents at the same time an essential way to deliver drugs and immune cells to the tumor. However, tumor vasculature exhibits many morphological and functional abnormalities, thus resulting in the formation of hypoxic areas within tumors, believed to represent a mechanism to maintain tumor cells in an invasive state.Tumors are vascularized through a variety of modalities, mainly represented by angiogenesis, where VEGF and other members of the VEGF family play a key role. This has represented the basis for the development of anti-VEGF blocking agents and their use in cancer therapy: however, these agents failed to induce significant therapeutic effects.Much less is known about the cellular origin of vessel network in tumors. Various cell types may contribute to tumor vasculature in different tumors or in the same tumor, such as mature endothelial cells, endothelial progenitor cells (EPCs), or the same tumor cells through a process of transdifferentiation. Early studies have suggested a role for bone marrow-derived EPCs; these cells do not are true EPCs but myeloid progenitors differentiating into monocytic cells, exerting a proangiogenic effect through a paracrine mechanism. More recent studies have shown the existence of tissue-resident endothelial vascular progenitors (EVPs) present at the level of vessel endothelium and their possible involvement as cells of origin of tumor vasculature.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, Rome, Italy.
| | - Elvira Pelosi
- Department of Oncology, Istituto Superiore di Sanità, Rome, Italy
| | - Germana Castelli
- Department of Oncology, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
33
|
Tian F, Wang H, Ma H, Zhong Y, Liao A. miR‑144‑3p inhibits the proliferation, migration and angiogenesis of multiple myeloma cells by targeting myocyte enhancer factor 2A. Int J Mol Med 2020; 46:1155-1165. [PMID: 32705159 PMCID: PMC7387096 DOI: 10.3892/ijmm.2020.4670] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 06/05/2020] [Indexed: 12/28/2022] Open
Abstract
Multiple myelomas (MM) are the second most common haematological malignancy, for which no curative treatments have been reported to date. MicroRNAs (miRNAs or miRs) have recently been shown to be involved in the proliferation of MM cells. However, the molecular mechanisms through which miRNAs regulate migration and angiogenesis in MM are poorly understood. Accordingly, the present study evaluated the role of miR-144-3p in MM. miR-144-3p exhibited a lower expression in patients with MM and in MM cell lines compared with normal cells (mononuclear cells derived from bone marrow). The transfection of miR-144-3p into MM cells inhibited proliferation, migration and angiogenesis, and induced cell cycle arrest and apoptosis compared to the control cells. Furthermore, miR-144-3p suppressed the transcription and translation of the myocyte enhancer factor 2A (MEF2A) gene and disrupted the expression of vascular endothelial growth factor. The knockdown of MEF2A significantly inhibited the proliferation, migration and angiogenesis of MM cells. However, the overexpression of MEF2A reversed these effects. On the whole, the findings of the present study demonstrate that miR-144-3p exerts antitumour effects by downregulating MEF2A to inhibit the proliferation, migration and angiogenesis of MM cells. This suggests that the miR-144-3p/MEF2A interaction may prove to be a potential therapeutic target for MM.
Collapse
Affiliation(s)
- Fei Tian
- Department of Haematology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Huihan Wang
- Department of Haematology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Huanxin Ma
- Department of Haematology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Yuan Zhong
- Department of Haematology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Aijun Liao
- Department of Haematology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
34
|
Ronca R, Ghedini GC, Maccarinelli F, Sacco A, Locatelli SL, Foglio E, Taranto S, Grillo E, Matarazzo S, Castelli R, Paganini G, Desantis V, Cattane N, Cattaneo A, Mor M, Carlo-Stella C, Belotti A, Roccaro AM, Presta M, Giacomini A. FGF Trapping Inhibits Multiple Myeloma Growth through c-Myc Degradation-Induced Mitochondrial Oxidative Stress. Cancer Res 2020; 80:2340-2354. [PMID: 32094301 DOI: 10.1158/0008-5472.can-19-2714] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/10/2019] [Accepted: 02/14/2020] [Indexed: 11/16/2022]
Abstract
Multiple myeloma, the second most common hematologic malignancy, frequently relapses because of chemotherapeutic resistance. Fibroblast growth factors (FGF) act as proangiogenic and mitogenic cytokines in multiple myeloma. Here, we demonstrate that the autocrine FGF/FGFR axis is essential for multiple myeloma cell survival and progression by protecting multiple myeloma cells from oxidative stress-induced apoptosis. In keeping with the hypothesis that the intracellular redox status can be a target for cancer therapy, FGF/FGFR blockade by FGF trapping or tyrosine kinase inhibitor impaired the growth and dissemination of multiple myeloma cells by inducing mitochondrial oxidative stress, DNA damage, and apoptotic cell death that were prevented by the antioxidant vitamin E or mitochondrial catalase overexpression. In addition, mitochondrial oxidative stress occurred as a consequence of proteasomal degradation of the c-Myc oncoprotein that led to glutathione depletion. Accordingly, expression of a proteasome-nondegradable c-Myc protein mutant was sufficient to avoid glutathione depletion and rescue the proapoptotic effects due to FGF blockade. These findings were confirmed on bortezomib-resistant multiple myeloma cells as well as on bone marrow-derived primary multiple myeloma cells from newly diagnosed and relapsed/refractory patients, including plasma cells bearing the t(4;14) translocation obtained from patients with high-risk multiple myeloma. Altogether, these findings dissect the mechanism by which the FGF/FGFR system plays a nonredundant role in multiple myeloma cell survival and disease progression, and indicate that FGF targeting may represent a therapeutic approach for patients with multiple myeloma with poor prognosis and advanced disease stage. SIGNIFICANCE: This study provides new insights into the mechanisms by which FGF antagonists promote multiple myeloma cell death. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/11/2340/F1.large.jpg.
Collapse
Affiliation(s)
- Roberto Ronca
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Gaia C Ghedini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Federica Maccarinelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Antonio Sacco
- Clinical Research Development and Phase I Unit, CREA Laboratory, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Silvia L Locatelli
- Department of Oncology and Hematology, Humanitas Clinical and Research Center - IRCCS and Humanitas University, Milan, Italy
| | - Eleonora Foglio
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Sara Taranto
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Elisabetta Grillo
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Sara Matarazzo
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | | - Giuseppe Paganini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Vanessa Desantis
- Department of Biomedical Sciences and Human Oncology, Azienda Ospedaliera Consorziale Universitaria Policlinico di Bari, Bari, Italy
| | - Nadia Cattane
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Annamaria Cattaneo
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.,Stress, Psychiatry and Immunology Laboratory, Department of Psychological Medicine, Institute of Psychiatry, King's College London, London, United Kingdom
| | - Marco Mor
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Carmelo Carlo-Stella
- Department of Oncology and Hematology, Humanitas Clinical and Research Center - IRCCS and Humanitas University, Milan, Italy
| | - Angelo Belotti
- Department of Hematology, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Aldo M Roccaro
- Clinical Research Development and Phase I Unit, CREA Laboratory, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Marco Presta
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| | - Arianna Giacomini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| |
Collapse
|
35
|
Bortezomib Treatment Modulates Autophagy in Multiple Myeloma. J Clin Med 2020; 9:jcm9020552. [PMID: 32085480 PMCID: PMC7073518 DOI: 10.3390/jcm9020552] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/12/2020] [Accepted: 02/13/2020] [Indexed: 12/11/2022] Open
Abstract
Although the introduction of bortezomib as a therapeutic strategy has improved the overall survival of multiple myeloma (MM) patients, 15–20% of high-risk patients do not respond to bortezomib over time or become resistant to treatment. Therefore, the development of new therapeutic strategies, such as combination therapies, is urgently needed. Methods: Given that bortezomib resistance may be mediated by activation of the autophagy pathway as an alternative mechanism of protein degradation, and that an enormous amounts of misfolded protein is generated in myeloma plasma cells (PCs), we investigated the effect of the simultaneous inhibition of proteasome by bortezomib and autophagy by hydroxychloroquine (HCQ) treatment on PCs and endothelial cells (ECs) isolated from patients with monoclonal gammopathy of undetermined significance (MGUS) and MM. Results: We found that bortezomib combined with HCQ induces synergistic cytotoxicity in myeloma PCs whereas this effect is lost on ECs. Levels of microtubule-associated protein light chain beta (LC3B) and p62 are differentially modulated in PCs and ECs, with effects on cell viability and proliferation. Conclusions: Our results suggest that treatment with bortezomib and HCQ should be associated with an anti-angiogenic drug to prevent the pro-angiogenic effect of bortezomib, the proliferation of a small residual tumor PC clone, and thus the relapse.
Collapse
|
36
|
Ciftci HI, Can M, Ellakwa DE, Suner SC, Ibrahim MA, Oral A, Sekeroglu N, Özalp B, Otsuka M, Fujita M, Alparslan M, Radwan MO. Anticancer activity of Turkish marine extracts: a purple sponge extract induces apoptosis with multitarget kinase inhibition activity. Invest New Drugs 2020; 38:1326-1333. [PMID: 32062733 DOI: 10.1007/s10637-020-00911-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 02/10/2020] [Indexed: 11/29/2022]
Abstract
Marine natural products have drawn a great deal of attention as a vital source of new drugs for the last five decades. However, marine organisms in the seas surrounding Turkey (the Black Sea, the Aegean Sea and the Mediterranean Sea) haven't been yet extensively explored. In the present study, three marine organisms (Dysidea avara, Microcosmus sabatieri and Echinaster sepositus) were sampled from the Dardanelles (Turkish Straits System, Western Turkey) by scientific divers, transferred to the laboratory and then were extracted with 70% ethanol. The extracts were tested for their cytotoxic effect against K562, KMS-12PE, A549, and A375 cancer cell lines. The sponge extract elicited the most promising cytotoxic activity, thus it was further evaluated against H929, MCF-7, HeLa, and HCT116 cancer cells. Most of the designated cells showed a considerable sensitivity for the sponge extract particularly H929, K562, KMS-12PE and HeLa cells with IC50 less than 10 μg/mL. On the contrary, the other two extracts exhibited no cytotoxic activity on all cells at 100 μg/mL concentration. The sponge extract was tested for its capacity to induce apoptosis in cancer cells and to inhibit a panel of tyrosine kinases showing remarkable results. The outcome of this study represents a platform for discovery of new chemotherapeutic agents of marine natural origin.
Collapse
Affiliation(s)
- Halil I Ciftci
- Department of Drug Discovery, Science Farm Ltd., 1-7-30-805 Kuhonji, Chuo-ku, Kumamoto, 862-0976, Japan.,Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-Ku, Kumamoto, 8620973, Japan
| | - Mustafa Can
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-Ku, Kumamoto, 8620973, Japan.,Department of Engineering Sciences, Faculty of Engineering and Architecture, Izmir Katip Celebi University, Havaalani Sosesi Caddesi No:25, 35620, Cigli/Izmir, Turkey
| | - Doha E Ellakwa
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-Ku, Kumamoto, 8620973, Japan.,Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Al-Azhar University, Greater Cairo, Nasr City, 11651, Egypt
| | - Salih C Suner
- Chemical and Chemical Processing, Vocational School of Lapseki, Canakkale Onsekiz Mart University, 17800, Canakkale, Turkey
| | - Mohamed A Ibrahim
- Chemistry of Natural Compounds Department, Pharmaceutical and Drug Industries Research Division, National Research Centre, Dokki, Cairo, 12622, Egypt
| | - Ayhan Oral
- Department of Chemistry, Faculty of Science, Canakkale Onsekiz Mart University, 17020, Canakkale, Turkey
| | - Nazim Sekeroglu
- Department of Horticulture, Faculty of Agriculture, Kilis 7 Aralik University, 79000, Kilis, Turkey
| | - Barış Özalp
- Section of Underwater Technology, Vocational School of Ocean Engineering, Canakkale Onsekiz Mart University, 17100, Canakkale, Turkey
| | - Masami Otsuka
- Department of Drug Discovery, Science Farm Ltd., 1-7-30-805 Kuhonji, Chuo-ku, Kumamoto, 862-0976, Japan.,Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-Ku, Kumamoto, 8620973, Japan
| | - Mikako Fujita
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-Ku, Kumamoto, 8620973, Japan.
| | - Mustafa Alparslan
- Faculty of Fisheries and Aquaculture, Izmir Katip Celebi University, 35620, Izmir, Turkey.
| | - Mohamed O Radwan
- Department of Drug Discovery, Science Farm Ltd., 1-7-30-805 Kuhonji, Chuo-ku, Kumamoto, 862-0976, Japan. .,Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-Ku, Kumamoto, 8620973, Japan. .,Chemistry of Natural Compounds Department, Pharmaceutical and Drug Industries Research Division, National Research Centre, Dokki, Cairo, 12622, Egypt.
| |
Collapse
|
37
|
Zhu FX, Wang XT, Ye ZZ, Gan ZP, Lai YR. Construction of a prognosis‑associated long noncoding RNA‑mRNA network for multiple myeloma based on microarray and bioinformatics analysis. Mol Med Rep 2020; 21:999-1010. [PMID: 32016443 PMCID: PMC7003030 DOI: 10.3892/mmr.2020.10930] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 04/10/2019] [Indexed: 02/07/2023] Open
Abstract
At present, the association between prognosis-associated long noncoding RNAs (lncRNAs) and mRNAs is yet to be reported in multiple myeloma (MM). The aim of the present study was to construct prognostic models with lncRNAs and mRNAs, and to map the interactions between these lncRNAs and mRNAs in MM. LncRNA and mRNA data from 559 patients with MM were acquired from the Genome Expression Omnibus (dataset GSE24080), and their prognostic values were calculated using the survival package in R. Multivariate Cox analysis was used on the top 20 most significant prognosis-associated mRNAs and lncRNAs to develop prognostic signatures. The performances of these prognostic signatures were tested using the survivalROC package in R, which allows for time-dependent receiver operator characteristic (ROC) curve estimation. Weighted correlation network analysis (WGCNA) was conducted to investigate the associations between lncRNAs and mRNAs, and a lncRNA-mRNA network was constructed using Cytoscape software. Univariate Cox regression analysis identified 39 lncRNAs and 1,445 mRNAs that were significantly associated with event-free survival of MM patients. The top 20 most significant survival-associated lncRNAs and mRNAs were selected as candidates for analyzing independent MM prognostic factors. Both signatures could be used to separate patients into two groups with distinct outcomes. The areas under the ROC curves were 0.739 for the lncRNA signature and 0.732 for the mRNA signature. In the lncRNA-mRNA network, a total of 143 mRNAs were positively or negatively associated with 23 prognosis-associated lncRNAs. NCRNA00201, LOC115110 and RP5-968J1.1 were the most dominant drivers. The present study constructed a model that predicted prognosis in MM and formed a network with the corresponding prognosis-associated mRNAs, providing a novel perspective for the clinical diagnosis and treatment of MM, and suggesting novel directions for interpreting the mechanisms underlying the development of MM.
Collapse
Affiliation(s)
- Fang-Xiao Zhu
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Xiao-Tao Wang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Zhi-Zhong Ye
- Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, Guangdong 518040, P.R. China
| | - Zhao-Ping Gan
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yong-Rong Lai
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
38
|
Mukherjee A, Madamsetty VS, Paul MK, Mukherjee S. Recent Advancements of Nanomedicine towards Antiangiogenic Therapy in Cancer. Int J Mol Sci 2020; 21:E455. [PMID: 31936832 PMCID: PMC7013812 DOI: 10.3390/ijms21020455] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 01/07/2020] [Accepted: 01/08/2020] [Indexed: 12/14/2022] Open
Abstract
Angiogenesis is a process of generation of de-novo blood vessels from already existing vasculature. It has a crucial role in different physiological process including wound healing, embryonic development, and tumor growth. The methods by which therapeutic drugs inhibit tumor angiogenesis are termed as anti-angiogenesis cancer therapy. Developments of angiogenic inhibiting drugs have various limitations causing a barrier for successful treatment of cancer, where angiogenesis plays an important role. In this context, investigators developed novel strategies using nanotechnological approaches that have demonstrated inherent antiangiogenic properties or used for the delivery of antiangiogenic agents in a targeted manner. In this present article, we decisively highlight the recent developments of various nanoparticles (NPs) including liposomes, lipid NPs, protein NPs, polymer NPs, inorganic NPs, viral and bio-inspired NPs for potential application in antiangiogenic cancer therapy. Additionally, the clinical perspectives, challenges of nanomedicine, and future perspectives are briefly analyzed.
Collapse
Affiliation(s)
- Anubhab Mukherjee
- Aavishkar Oral Strips Pvt Ltd., 109/3, IDA, Phase 2, Sector 2, Lane 6, Cherlapally, Hyderabad 500051, India;
| | - Vijay Sagar Madamsetty
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA;
| | - Manash K. Paul
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, The University of California, Los Angeles (UCLA), Factor Bldg. 10-240, 621 Charles E. Young Dr., Los Angeles, CA 90095, USA
| | - Sudip Mukherjee
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| |
Collapse
|
39
|
Yang Q, Li K, Li X, Liu J. Identification of Key Genes and Pathways in Myeloma side population cells by Bioinformatics Analysis. Int J Med Sci 2020; 17:2063-2076. [PMID: 32922167 PMCID: PMC7484674 DOI: 10.7150/ijms.48244] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 07/12/2020] [Indexed: 12/17/2022] Open
Abstract
Background: Multiple myeloma (MM) is the second most common hematological malignancy, which is still incurable and relapses inevitably, highlighting further understanding of the possible mechanisms. Side population (SP) cells are a group of enriched progenitor cells showing stem-like phenotypes with a distinct low-staining pattern with Hoechst 33342. Compared to main population (MP) cells, the underlying molecular characteristics of SP cells remain largely unclear. This bioinformatics analysis aimed to identify key genes and pathways in myeloma SP cells to provide novel biomarkers, predict MM prognosis and advance potential therapeutic targets. Methods: The gene expression profile GSE109651 was obtained from Gene Expression Omnibus database, and then differentially expressed genes (DEGs) with P-value <0.05 and |log2 fold-change (FC)| > 2 were selected by the comparison of myeloma light-chain (LC) restricted SP (LC/SP) cells and MP CD138+ cells. Subsequently, gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) pathway enrichment analysis, protein-protein interaction (PPI) network analysis were performed to identify the functional enrichment analysis of the DEGs and screen hub genes. Cox proportional hazards regression was used to select the potential prognostic DEGs in training dataset (GSE2658). The prognostic value of the potential prognostic genes was evaluated by Kaplan-Meier curve and validated in another external dataset (MMRF-CoMMpass cohort from TCGA). Results: Altogether, 403 up-regulated and 393 down-regulated DEGs were identified. GO analysis showed that the up-regulated DEGs were significantly enriched in innate immune response, inflammatory response, plasma membrane and integral component of membrane, while the down-regulated DEGs were mainly involved in protoporphyrinogen IX and heme biosynthetic process, hemoglobin complex and erythrocyte differentiation. KEGG pathway analysis suggested that the DEGs were significantly enriched in osteoclast differentiation, porphyrin and chlorophyll metabolism and cytokine-cytokine receptor interaction. The top 10 hub genes, identified by the plug-in cytoHubba of the Cytoscape software using maximal clique centrality (MCC) algorithm, were ITGAM, MMP9, ITGB2, FPR2, C3AR1, CXCL1, CYBB, LILRB2, HP and FCER1G. Modules and corresponding GO enrichment analysis indicated that myeloma LC/SP cells were significantly associated with immune system, immune response and cell cycle. The predictive value of the prognostic model including TFF3, EPDR1, MACROD1, ARHGEF12, AMMECR1, NFATC2, HES6, PLEK2 and SNCA was identified, and validated in another external dataset (MMRF-CoMMpass cohort from TCGA). Conclusions: In conclusion, this study provides reliable molecular biomarkers for screening, prognosis, as well as novel therapeutic targets for myeloma LC/SP cells.
Collapse
Affiliation(s)
- Qin Yang
- Department of Hematology, the Third Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Kaihu Li
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Xin Li
- Department of Hematology, the Third Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Jing Liu
- Department of Hematology, the Third Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| |
Collapse
|
40
|
Pacini S, Montali M, Mazziotta F, Schifone CP, Macchia L, Carnicelli V, Panvini FM, Barachini S, Notarfranchi L, Previti GB, Buda G, Petrini M. Mesangiogenic progenitor cells are forced toward the angiogenic fate, in multiple myeloma. Oncotarget 2019; 10:6781-6790. [PMID: 31827721 PMCID: PMC6887577 DOI: 10.18632/oncotarget.27285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 10/04/2019] [Indexed: 11/25/2022] Open
Abstract
Multiple myeloma (MM) progresses mainly in the bone marrow where the involvement of a specific microenvironment plays a critical role in maintaining plasma cell growth, spread, and survival. In active disease, the switch from a pre-vascular/non-active phase to a vascular phase is coupled with the impairment of bone turnover. Previously, we have isolated Mesangiogenic Progenitor Cells (MPCs), a bone marrow population that showed mesengenic and angiogenic potential, both in vitro and in vivo. MPC differentiation into musculoskeletal tissue and their ability of sprouting angiogenesis are mutually exclusive, suggesting a role in the imbalancing of the microenvironment in multiple myeloma. MPCs from 32 bone marrow samples of multiple myeloma and 23 non-hematological patients were compared in terms of frequency, phenotype, mesengenic/angiogenic potential, and gene expression profile. Defective osteogenesis was recorded for MM-derived MPCs that showed longer angiogenic sprouting distances respect to non-hematological MPCs, retaining this capability after mesengenic induction. This altered MPCs differentiation potential was not detected in asymptomatic myelomatous disease. These in vitro experiments are suggestive of a forced angiogenic fate in MPCs isolated from MM patients, which also showed increased sprouting activity. Taking together our results suggest a possible role of these cells in the “angiogenic switch” in the MM micro-environment.
Collapse
Affiliation(s)
- Simone Pacini
- Department of Clinical and Experimental Medicine, Hematology Division, University of Pisa, Pisa, Italy
| | - Marina Montali
- Department of Clinical and Experimental Medicine, Hematology Division, University of Pisa, Pisa, Italy
| | | | - Claudia P Schifone
- Department of Clinical and Experimental Medicine, Hematology Division, University of Pisa, Pisa, Italy
| | - Lucia Macchia
- Department of Laboratory Medicine, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Vittoria Carnicelli
- Department of Surgical, Medical, and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Francesca M Panvini
- Institute of Life Sciences, Sant'Anna School of Advanced Studies, Pisa, Italy
| | - Serena Barachini
- Department of Clinical and Experimental Medicine, Hematology Division, University of Pisa, Pisa, Italy
| | - Laura Notarfranchi
- Department of Medicine and Surgery, Hematology Division, University of Parma, Parma, Italy
| | | | - Gabriele Buda
- Department of Clinical and Experimental Medicine, Hematology Division, University of Pisa, Pisa, Italy
| | - Mario Petrini
- Department of Clinical and Experimental Medicine, Hematology Division, University of Pisa, Pisa, Italy
| |
Collapse
|
41
|
Melegh Z, Oltean S. Targeting Angiogenesis in Prostate Cancer. Int J Mol Sci 2019; 20:E2676. [PMID: 31151317 PMCID: PMC6600172 DOI: 10.3390/ijms20112676] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 05/24/2019] [Accepted: 05/29/2019] [Indexed: 02/07/2023] Open
Abstract
Prostate cancer is the most commonly diagnosed cancer among men in the Western world. Although localized disease can be effectively treated with established surgical and radiopharmaceutical treatments options, the prognosis of castration-resistant advanced prostate cancer is still disappointing. The objective of this study was to review the role of angiogenesis in prostate cancer and to investigate the effectiveness of anti-angiogenic therapies. A literature search of clinical trials testing the efficacy of anti-angiogenic therapy in prostate cancer was performed using Pubmed. Surrogate markers of angiogenic activity (microvessel density and vascular endothelial growth factor A (VEGF-A) expression) were found to be associated with tumor grade, metastasis, and prognosis. Six randomizedstudies were included in this review: two phase II trials on localized and hormone-sensitive disease (n = 60 and 99 patients) and four phase III trials on castration-resistant refractory disease (n = 873 to 1224 patients). Although the phase II trials showed improved relapse-free survival and stabilisation of the disease, the phase III trials found increased toxicity and no significant improvement in overall survival. Although angiogenesis appears to have an important role in prostate cancer, the results of anti-angiogenic therapy in castration-resistant refractory disease have hitherto been disappointing. There are various possible explanations for this lack of efficacy in castration-resistant refractory disease: redundancy of angiogenic pathways, molecular heterogeneity of the disease, loss of tumor suppressor protein phosphatase and tensin homolog (PTEN) expression as well as various VEGF-A splicing isoforms with pro- and anti-angiogenic activity. A better understanding of the molecular mechanisms of angiogenesis may help to develop effective anti-angiogenic therapy in prostate cancer.
Collapse
Affiliation(s)
- Zsombor Melegh
- Department of Cellular Pathology, Southmead Hospital, Bristol BS10 5NB, UK.
| | - Sebastian Oltean
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, Exeter EX12LU, UK.
| |
Collapse
|
42
|
Berlier JL, Rethnam M, Banu Binte Abdul Majeed A, Suda T. Modification of the bone marrow MSC population in a xenograft model of early multiple myeloma. Biochem Biophys Res Commun 2019; 508:1175-1181. [DOI: 10.1016/j.bbrc.2018.11.178] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 11/27/2018] [Indexed: 01/21/2023]
|