1
|
Culkins C, Adomanis R, Phan N, Robinson B, Slaton E, Lothrop E, Chen Y, Kimmel BR. Unlocking the Gates: Therapeutic Agents for Noninvasive Drug Delivery Across the Blood-Brain Barrier. Mol Pharm 2024. [PMID: 39324552 DOI: 10.1021/acs.molpharmaceut.4c00604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
The blood-brain barrier (BBB) is a highly selective network of various cell types that acts as a filter between the blood and the brain parenchyma. Because of this, the BBB remains a major obstacle for drug delivery to the central nervous system (CNS). In recent years, there has been a focus on developing various modifiable platforms, such as monoclonal antibodies (mAbs), nanobodies (Nbs), peptides, and nanoparticles, as both therapeutic agents and carriers for targeted drug delivery to treat brain cancers and diseases. Methods for bypassing the BBB can be invasive or noninvasive. Invasive techniques, such as transient disruption of the BBB using low pulse electrical fields and intracerebroventricular infusion, lack specificity and have numerous safety concerns. In this review, we will focus on noninvasive transport mechanisms that offer high levels of biocompatibility, personalization, specificity and are regarded as generally safer than their invasive counterparts. Modifiable platforms can be designed to noninvasively traverse the BBB through one or more of the following pathways: passive diffusion through a physio-pathologically disrupted BBB, adsorptive-mediated transcytosis, receptor-mediated transcytosis, shuttle-mediated transcytosis, and somatic gene transfer. Through understanding the noninvasive pathways, new applications, including Chimeric Antigen Receptors T-cell (CAR-T) therapy, and approaches for drug delivery across the BBB are emerging.
Collapse
Affiliation(s)
- Courtney Culkins
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Roman Adomanis
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Nathan Phan
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Blaise Robinson
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Ethan Slaton
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Elijah Lothrop
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Yinuo Chen
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Blaise R Kimmel
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
- Center for Cancer Engineering, Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
- Pelotonia Institute for Immuno-Oncology, Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
2
|
Lu K, Wang W, Liu Y, Xie C, Liu J, Xing L. Advancements in microenvironment-based therapies: transforming the landscape of multiple myeloma treatment. Front Oncol 2024; 14:1413494. [PMID: 39087026 PMCID: PMC11288838 DOI: 10.3389/fonc.2024.1413494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 06/20/2024] [Indexed: 08/02/2024] Open
Abstract
Multiple myeloma (MM) is the most prevalent malignant monoclonal disease of plasma cells. There is mounting evidence that interactions with the bone marrow (BM) niche are essential for the differentiation, proliferation, survival, migration, and treatment resistance of myeloma cells. For this reason, gaining a deeper comprehension of how BM microenvironment compartments interact with myeloma cells may inspire new therapeutic ideas that enhance patient outcomes. This review will concentrate on the most recent findings regarding the mechanisms of interaction between microenvironment and MM and highlight research on treatment targeting the BM niche.
Collapse
Affiliation(s)
- Ke Lu
- Department of Lymphoma, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Wen Wang
- Department of Lymphoma, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yuntong Liu
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - Chao Xie
- Department of Respiratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jiye Liu
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - Lijie Xing
- Department of Lymphoma, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Key Laboratory of Biopharmaceuticals, Postdoctoral Scientific Research Workstation, Shandong Academy of Pharmaceutical Science, Jinan, Shandong, China
| |
Collapse
|
3
|
Su MC, Nethi SK, Dhanyamraju PK, Prabha S. Nanomedicine Strategies for Targeting Tumor Stroma. Cancers (Basel) 2023; 15:4145. [PMID: 37627173 PMCID: PMC10452920 DOI: 10.3390/cancers15164145] [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: 07/19/2023] [Revised: 08/04/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
The tumor stroma, or the microenvironment surrounding solid tumors, can significantly impact the effectiveness of cancer therapies. The tumor microenvironment is characterized by high interstitial pressure, a consequence of leaky vasculature, and dense stroma created by excessive deposition of various macromolecules such as collagen, fibronectin, and hyaluronic acid (HA). In addition, non-cancerous cells such as cancer-associated fibroblasts (CAFs) and the extracellular matrix (ECM) itself can promote tumor growth. In recent years, there has been increased interest in combining standard cancer treatments with stromal-targeting strategies or stromal modulators to improve therapeutic outcomes. Furthermore, the use of nanomedicine, which can improve the delivery and retention of drugs in the tumor, has been proposed to target the stroma. This review focuses on how different stromal components contribute to tumor progression and impede chemotherapeutic delivery. Additionally, this review highlights recent advancements in nanomedicine-based stromal modulation and discusses potential future directions for developing more effective stroma-targeted cancer therapies.
Collapse
Affiliation(s)
- Mei-Chi Su
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Susheel Kumar Nethi
- Nanovaccine Institute, Department of Chemical & Biological Engineering, Iowa State University, Ames, IA 50011, USA;
| | - Pavan Kumar Dhanyamraju
- Fels Cancer Institute of Personalized Medicine, Lewis-Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA;
| | - Swayam Prabha
- Fels Cancer Institute of Personalized Medicine, Lewis-Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA;
- Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Temple University, Philadelphia, PA 19111, USA
| |
Collapse
|
4
|
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
|
5
|
Past, Present, and a Glance into the Future of Multiple Myeloma Treatment. Pharmaceuticals (Basel) 2023; 16:ph16030415. [PMID: 36986514 PMCID: PMC10056051 DOI: 10.3390/ph16030415] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/11/2023] Open
Abstract
Multiple myeloma (MM) is a challenging hematological cancer which typically grows in bone marrow. MM accounts for 10% of hematological malignancies and 1.8% of cancers. The recent treatment strategies have significantly improved progression-free survival for MM patients in the last decade; however, a relapse for most MM patients is inevitable. In this review we discuss current treatment, important pathways for proliferation, survival, immune suppression, and resistance that could be targeted for future treatments.
Collapse
|
6
|
Periplocin Overcomes Bortezomib Resistance by Suppressing the Growth and Down-Regulation of Cell Adhesion Molecules in Multiple Myeloma. Cancers (Basel) 2023; 15:cancers15051526. [PMID: 36900317 PMCID: PMC10001131 DOI: 10.3390/cancers15051526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/17/2023] [Accepted: 02/21/2023] [Indexed: 03/05/2023] Open
Abstract
Multiple myeloma (MM) is an incurable hematological malignant disorder of bone marrow. Patients with MM receive multiple lines of chemotherapeutic treatments which often develop bortezomib (BTZ) resistance and relapse. Therefore, it is crucial to identify an anti-MM agent to overcome the BTZ resistance of MM. In this study, we screened a library of 2370 compounds against MM wild-type (ARP1) and BTZ-resistant type (ARP1-BR) cell lines and found that periplocin (PP) was the most significant anti-MM natural compound. We further investigated the anti-MM effect of PP by using annexin V assay, clonogenic assays, aldefluor assay, and transwell assay. Furthermore, RNA sequencing (RNA-seq) was performed to predict the molecular effects of PP in MM followed by verification through qRT-PCR and Western blot analysis. Moreover, ARP1 and ARP1-BR xenograft mice models of MM were established to confirm the anti-MM effects of PP invivo. The results showed that PP significantly induced apoptosis, inhibited proliferation, suppressed stemness, and reduced the cell migration of MM. The expression of cell adhesion molecules (CAMs) was suppressed upon PP treatment in vitro and in vivo. Overall, our data recommend PP as an anti-MM natural compound with the potential to overcome BTZ resistance and downregulate CAMs in MM.
Collapse
|
7
|
Johnson CS, Cook LM. Osteoid cell-derived chemokines drive bone-metastatic prostate cancer. Front Oncol 2023; 13:1100585. [PMID: 37025604 PMCID: PMC10070788 DOI: 10.3389/fonc.2023.1100585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 03/07/2023] [Indexed: 04/08/2023] Open
Abstract
One of the greatest challenges in improving prostate cancer (PCa) survival is in designing new therapies to effectively target bone metastases. PCa regulation of the bone environment has been well characterized; however, bone-targeted therapies have little impact on patient survival, demonstrating a need for understanding the complexities of the tumor-bone environment. Many factors contribute to creating a favorable microenvironment for prostate tumors in bone, including cell signaling proteins produced by osteoid cells. Specifically, there has been extensive evidence from both past and recent studies that emphasize the importance of chemokine signaling in promoting PCa progression in the bone environment. Chemokine-focused strategies present promising therapeutic options for treating bone metastasis. These signaling pathways are complex, with many being produced by (and exerting effects on) a plethora of different cell types, including stromal and tumor cells of the prostate tumor-bone microenvironment. This review highlights an underappreciated molecular family that should be interrogated for treatment of bone metastatic prostate cancer (BM-PCa).
Collapse
Affiliation(s)
- Catherine S. Johnson
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
- Eppley Institute for Research in Cancer and Allied Diseases, Omaha, NE, United States
| | - Leah M. Cook
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States
- *Correspondence: Leah M. Cook,
| |
Collapse
|
8
|
Liao Y, Xiong S, Ur Rehman Z, He X, Peng H, Liu J, Sun S. The Research Advances of Aptamers in Hematologic Malignancies. Cancers (Basel) 2023; 15:300. [PMID: 36612296 PMCID: PMC9818631 DOI: 10.3390/cancers15010300] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/28/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
Currently, research for hematological malignancies is very intensive, with many breakthroughs. Among them, aptamer-based targeted therapies could be counted. Aptamer is a targeting tool with many unique advantages (easy synthesis, low toxicity, easy modification, low immunogenicity, nano size, long stability, etc.), therefore many experts screened corresponding aptamers in various hematological malignancies for diagnosis and treatment. In this review, we try to summarize and provide the recent progress of aptamer research in the diagnosis and treatment of hematologic malignancies. Until now, 29 aptamer studies were reported in hematologic malignancies, of which 12 aptamers were tested in vivo and the remaining 17 aptamers were only tested in vitro. In this case, 11 aptamers were combined with chemotherapeutic drugs for the treatment of hematologic malignancies, 4 aptamers were used in combination with nanomaterials for the diagnosis and treatment of hematologic malignancies, and some studies used aptamers for the targeted transportation of siRNA and miRNA for targeted therapeutic effects. Their research provides multiple approaches to achieve more targeted goals. These findings show promising and encouraging future for both hematological malignancies basic and clinical trials research.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shuming Sun
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, Center for Medical Genetics, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China
| |
Collapse
|
9
|
Amundarain A, Pastor F, Prósper F, Agirre X. Aptamers, a New Therapeutic Opportunity for the Treatment of Multiple Myeloma. Cancers (Basel) 2022; 14:5471. [PMID: 36358889 PMCID: PMC9657029 DOI: 10.3390/cancers14215471] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/31/2022] [Accepted: 11/04/2022] [Indexed: 08/30/2023] Open
Abstract
Multiple Myeloma (MM) remains an incurable disease due to high relapse rates and fast development of drug resistances. The introduction of monoclonal antibodies (mAb) has caused a paradigm shift in MM treatment, paving the way for targeted approaches with increased efficacy and reduced toxicities. Nevertheless, antibody-based therapies face several difficulties such as high immunogenicity, high production costs and limited conjugation capacity, which we believe could be overcome by the introduction of nucleic acid aptamers. Similar to antibodies, aptamers can bind to their targets with great affinity and specificity. However, their chemical nature reduces their immunogenicity and production costs, while it enables their conjugation to a wide variety of cargoes for their use as delivery agents. In this review, we summarize several aptamers that have been tested against MM specific targets with promising results, establishing the rationale for the further development of aptamer-based strategies against MM. In this direction, we believe that the study of novel plasma cell surface markers, the development of intracellular aptamers and further research on aptamers as building blocks for complex nanomedicines will lead to the generation of next-generation targeted approaches that will undoubtedly contribute to improve the management and life quality of MM patients.
Collapse
Affiliation(s)
- Ane Amundarain
- Center for Applied Medical Research (CIMA), IDISNA, University of Navarra, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 31008 Pamplona, Spain
| | - Fernando Pastor
- Center for Applied Medical Research (CIMA), IDISNA, University of Navarra, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 31008 Pamplona, Spain
| | - Felipe Prósper
- Center for Applied Medical Research (CIMA), IDISNA, University of Navarra, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 31008 Pamplona, Spain
- Hematology Department, Clínica Universidad de Navarra, CCUN, University of Navarra, 31008 Pamplona, Spain
| | - Xabier Agirre
- Center for Applied Medical Research (CIMA), IDISNA, University of Navarra, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 31008 Pamplona, Spain
| |
Collapse
|
10
|
Dadzie TG, Green AC. The role of the bone microenvironment in regulating myeloma residual disease and treatment. Front Oncol 2022; 12:999939. [PMID: 36072809 PMCID: PMC9441696 DOI: 10.3389/fonc.2022.999939] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/08/2022] [Indexed: 11/28/2022] Open
Abstract
Multiple myeloma is an incurable haematological cancer. The increase in targeted therapies has improved the number of myeloma patients achieving a complete response and improved progression-free survival following therapy. However, a low level of disease or minimal residual disease (MRD) still persists which contributes to the inevitable relapse in myeloma patients. MRD has been attributed to the presence of dormant myeloma cells and their subsequent reactivation, which is controlled by the microenvironment and specialised niches within the bone marrow. This contributes to the evasion of the immune system and chemotherapy, eventually leading to relapse. The growth of myeloma tumours are heavily dependent on environmental stimuli from the bone marrow microenvironment, and this plays a key role in myeloma progression. The bone microenvironment also plays a critical role in myeloma bone disease and the development of skeletal-related events. This review focuses on the bone marrow microenvironment in relation to myeloma pathogenesis and cancer dormancy. Moreover, it reviews the current therapies targeting the bone microenvironment to treat myeloma and myeloma bone disease. Lastly, it identifies novel therapeutic targets for myeloma treatment and the associated bone disease.
Collapse
|
11
|
Integrin Signaling Shaping BTK-Inhibitor Resistance. Cells 2022; 11:cells11142235. [PMID: 35883678 PMCID: PMC9322986 DOI: 10.3390/cells11142235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/12/2022] [Accepted: 07/12/2022] [Indexed: 11/16/2022] Open
Abstract
Integrins are adhesion molecules that function as anchors in retaining tumor cells in supportive tissues and facilitating metastasis. Beta1 integrins are known to contribute to cell adhesion-mediated drug resistance in cancer. Very late antigen-4 (VLA-4), a CD49d/CD29 heterodimer, is a beta1 integrin implicated in therapy resistance in both solid tumors and haematological malignancies such as chronic lymphocytic leukemia (CLL). A complex inside-out signaling mechanism activates VLA-4, which might include several therapeutic targets for CLL. Treatment regimens for this disease have recently shifted towards novel agents targeting BCR signaling. Bruton’s tyrosine kinase (BTK) is a component of B cell receptor signaling and BTK inhibitors such as ibrutinib are highly successful; however, their limitations include indefinite drug administration, the development of therapy resistance, and toxicities. VLA-4 might be activated independently of BTK, resulting in an ongoing interaction of CD49d-expressing leukemic cells with their surrounding tissue, which may reduce the success of therapy with BTK inhibitors and increases the need for alternative therapies. In this context, we discuss the inside-out signaling cascade culminating in VLA-4 activation, consider the advantages and disadvantages of BTK inhibitors in CLL and elucidate the mechanisms behind cell adhesion-mediated drug resistance.
Collapse
|
12
|
A review on the therapeutic applications of aptamers and aptamer-conjugated nanoparticles in cancer, inflammatory and viral diseases. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2021.103626] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
13
|
Waldschmidt JM, Fruttiger SJ, Wider D, Jung J, Thomsen AR, Hartmann TN, Duyster J, Hug MJ, Azab KA, Jung M, Wäsch R, Engelhardt M. Ex vivo propagation in a novel 3D high-throughput co-culture system for multiple myeloma. J Cancer Res Clin Oncol 2022; 148:1045-1055. [PMID: 35072775 PMCID: PMC9016043 DOI: 10.1007/s00432-021-03854-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 11/04/2021] [Indexed: 12/28/2022]
Abstract
Purpose Multiple myeloma (MM) remains an incurable hematologic malignancy which ultimately develops drug resistance and evades treatment. Despite substantial therapeutic advances over the past years, the clinical failure rate of preclinically promising anti-MM drugs remains substantial. More realistic in vitro models are thus required to better predict clinical efficacy of a preclinically active compound. Methods Here, we report on the establishment of a conical agarose 3D co-culture platform for the preclinical propagation of primary MM cells ex vivo. Cell growth was compared to yet established 2D and liquid overlay systems. MM cell lines (MMCL: RPMI-8226, U266, OPM-2) and primary patient specimens were tested. Drug sensitivity was examined by exploring the cytotoxic effect of bortezomib and the deubiquitinase inhibitor auranofin under various conditions. Results In contrast to 2D and liquid overlay, cell proliferation in the 3D array followed a sigmoidal curve characterized by an initial growth delay but more durable proliferation of MMCL over 12 days of culture. Primary MM specimens did not expand in ex vivo monoculture, but required co-culture support by a human stromal cell line (HS-5, MSP-1). HS-5 induced a > fivefold increase in cluster volume and maintained long-term viability of primary MM cells for up to 21 days. Bortezomib and auranofin induced less cytotoxicity under 3D vs. 2D condition and in co- vs. monoculture, respectively. Conclusions This study introduces a novel model that is capable of long-term propagation and drug testing of primary MM specimens ex vivo overcoming some of the pitfalls of currently available in vitro models. Supplementary Information The online version contains supplementary material available at 10.1007/s00432-021-03854-6.
Collapse
Affiliation(s)
- Johannes M Waldschmidt
- Department of Internal Medicine I, Faculty of Medicine and Medical Center, University of Freiburg, Hugstetterstr. 53, 79106, Freiburg, Germany
- Comprehensive Cancer Center Freiburg (CCCF), Freiburg University Medical Center, Freiburg, Germany
| | - Stefan J Fruttiger
- Department of Internal Medicine I, Faculty of Medicine and Medical Center, University of Freiburg, Hugstetterstr. 53, 79106, Freiburg, Germany
- Pharmacy, Freiburg University Medical Center, Freiburg, Germany
| | - Dagmar Wider
- Department of Internal Medicine I, Faculty of Medicine and Medical Center, University of Freiburg, Hugstetterstr. 53, 79106, Freiburg, Germany
| | - Johannes Jung
- Department of Internal Medicine I, Faculty of Medicine and Medical Center, University of Freiburg, Hugstetterstr. 53, 79106, Freiburg, Germany
- Comprehensive Cancer Center Freiburg (CCCF), Freiburg University Medical Center, Freiburg, Germany
| | - Andreas R Thomsen
- Department of Radiation Oncology, Freiburg University Medical Center, Freiburg, Germany
| | - Tanja N Hartmann
- Department of Internal Medicine I, Faculty of Medicine and Medical Center, University of Freiburg, Hugstetterstr. 53, 79106, Freiburg, Germany
- Comprehensive Cancer Center Freiburg (CCCF), Freiburg University Medical Center, Freiburg, Germany
| | - Justus Duyster
- Department of Internal Medicine I, Faculty of Medicine and Medical Center, University of Freiburg, Hugstetterstr. 53, 79106, Freiburg, Germany
- Comprehensive Cancer Center Freiburg (CCCF), Freiburg University Medical Center, Freiburg, Germany
| | - Martin J Hug
- Pharmacy, Freiburg University Medical Center, Freiburg, Germany
| | - Kareem A Azab
- Department of Radiation Oncology, Washington University, St. Louis, MO, USA
| | - Manfred Jung
- Institute of Pharmaceutical Sciences, University of Freiburg, Freiburg, Germany
| | - Ralph Wäsch
- Department of Internal Medicine I, Faculty of Medicine and Medical Center, University of Freiburg, Hugstetterstr. 53, 79106, Freiburg, Germany
- Comprehensive Cancer Center Freiburg (CCCF), Freiburg University Medical Center, Freiburg, Germany
| | - Monika Engelhardt
- Department of Internal Medicine I, Faculty of Medicine and Medical Center, University of Freiburg, Hugstetterstr. 53, 79106, Freiburg, Germany.
- Comprehensive Cancer Center Freiburg (CCCF), Freiburg University Medical Center, Freiburg, Germany.
| |
Collapse
|
14
|
Huang Y, Wang Y, Tang J, Qin S, Shen X, He S, Ju S. CAM-DR: Mechanisms, Roles and Clinical Application in Tumors. Front Cell Dev Biol 2021; 9:698047. [PMID: 34295898 PMCID: PMC8290360 DOI: 10.3389/fcell.2021.698047] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/08/2021] [Indexed: 12/14/2022] Open
Abstract
Despite the continuous improvement of various therapeutic techniques, the overall prognosis of tumors has been significantly improved, but malignant tumors in the middle and advanced stages still cannot be completely cured. It is now evident that cell adhesion-mediated resistance (CAM-DR) limits the success of cancer therapies and is a great obstacle to overcome in the clinic. The interactions between tumor cells and extracellular matrix (ECM) molecules or adjacent cells may play a significant role in initiating the intracellular signaling pathways that are associated with cell proliferation, survival upon binding to their ligands. Recent studies illustrate that these adhesion-related factors may contribute to the survival of cancer cells after chemotherapeutic therapy, advantageous to resistant cells to proliferate and develop multiple mechanisms of drug resistance. In this review, we focus on the molecular basis of these interactions and the main signal transduction pathways that are involved in the enhancement of the cancer cells’ survival. Furthermore, therapies targeting interactions between cancer cells and their environment to enhance drug response or prevent the emergence of drug resistance will also be discussed.
Collapse
Affiliation(s)
- Yuejiao Huang
- Medical School, Nantong University, Nantong, China.,Department of Medical Oncology, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Yuchan Wang
- Department of Pathogenic Biology, School of Medicine, Nantong University, Nantong, China
| | - Jie Tang
- Department of Pathogenic Biology, School of Medicine, Nantong University, Nantong, China
| | - Shiyi Qin
- Medical School, Nantong University, Nantong, China.,Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Xianjuan Shen
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Song He
- Department of Pathology, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Shaoqing Ju
- Medical School, Nantong University, Nantong, China.,Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
15
|
Solimando AG, Da Vià MC, Leone P, Borrelli P, Croci GA, Tabares P, Brandl A, Di Lernia G, Bianchi FP, Tafuri S, Steinbrunn T, Balduini A, Melaccio A, De Summa S, Argentiero A, Rauert-Wunderlich H, Frassanito MA, Ditonno P, Henke E, Klapper W, Ria R, Terragna C, Rasche L, Rosenwald A, Kortüm MK, Cavo M, Ribatti D, Racanelli V, Einsele H, Vacca A, Beilhack A. Halting the vicious cycle within the multiple myeloma ecosystem: blocking JAM-A on bone marrow endothelial cells restores angiogenic homeostasis and suppresses tumor progression. Haematologica 2021; 106:1943-1956. [PMID: 32354870 PMCID: PMC8252928 DOI: 10.3324/haematol.2019.239913] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Indexed: 12/26/2022] Open
Abstract
Interactions of malignant multiple myeloma (MM) plasma cells with the microenvironment control MM plasma-cell growth, survival, drug-resistance and dissemination. As microvascular density increases in the bone marrow in MM, we investigated whether bone marrow MM endothelial cells control disease progression via the junctional adhesion molecule-A (JAM-A). Membrane and cytoplasmic JAM-A levels were upregulated in MM endothelial cells in 111 patients with newly diagnosed MM and in 201 with relapsed/refractory MM compared to the levels in patients with monoclonal gammopathy of undetermined significance and healthy controls. Elevated membrane expression of JAM-A on MM endothelial cells predicted poor clinical outcome. Mechanistically, addition of recombinant JAM-A to MM endothelial cells increased angiogenesis, whereas inhibition of this adhesion molecule impaired angiogenesis and MM growth in two-dimensional and three-dimensional in vitro cell cultures and chorioallantoic membrane assays. To corroborate these findings, we treated MM-bearing mice with a JAM-A-blocking monoclonal antibody and demonstrated impaired MM progression, corresponding to decreased MM-related vascularity. These findings support the concept that JAM-A is an important mediator of MM progression through facilitating MM-associated angiogenesis. Elevated JAM-A expression on bone marrow endothelial cells is an independent prognostic factor for the survival of both patients with newly diagnosed MM and those with relapsed/refractory MM. Blocking JAM-A restricts angiogenesis in vitro, in utero and in vivo and represents a suitable druggable molecule to halt neo-angiogenesis and MM progression.
Collapse
Affiliation(s)
- Antonio G Solimando
- Department of Medicine II, University Hospital of Würzburg, Würzburg, Germany; 2IRCCS Istituto Tumori Giovanni Paolo II of Bari, Italy; 3University of Bari Aldo Moro Medical School, Bari, Italy
| | - Matteo C Da Vià
- Department of Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Patrizia Leone
- University of Bari Aldo Moro Medical School, Bari, Italy
| | - Paola Borrelli
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy
| | - Giorgio A Croci
- Department of Pathology, University of Kiel/University Hospital Schleswig-Holstein, Kiel, Germany; Department of Pathophysiology and Transplantation, University of Milan and Fondazione IRCCS, Ca' Granda, Milan, Italy
| | - Paula Tabares
- Department of Medicine II, University Hospital of Würzburg, Würzburg, Germany; 7Interdisciplinary Center for Clinical Research Laboratory, University Hospital of Würzburg, Würzburg, Germany
| | - Andreas Brandl
- Department of Medicine II, University Hospital of Würzburg, Würzburg, Germany; 7Interdisciplinary Center for Clinical Research Laboratory, University Hospital of Würzburg, Würzburg, Germany
| | | | - Francesco P Bianchi
- Department of Biomedical Science and Human Oncology, University of Aldo Moro Medical School, Bari, Italy
| | - Silvio Tafuri
- Department of Biomedical Science and Human Oncology, University of Aldo Moro Medical School, Bari, Italy
| | - Torsten Steinbrunn
- Department of Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy 10Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | | | - Simona De Summa
- Molecular Diagnostics and Pharmacogenetics Unit, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| | | | | | | | | | - Erik Henke
- Institute of Anatomy and Cell Biology, Julius-Maximilians Universität Würzburg, Würzburg, Germany
| | - Wolfram Klapper
- Department of Pathology, University of Kiel/University Hospital Schleswig-Holstein, Kiel, Germany
| | - Roberto Ria
- University of Bari Aldo Moro Medical School, Bari, Italy
| | | | - Leo Rasche
- Department of Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | | | - Martin K Kortüm
- Department of Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Michele Cavo
- Institute of Hematology L. and A. Seràgnoli, Bologna, Italy
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Aldo Moro Medical School, Bari, Italy
| | - Vito Racanelli
- University of Bari Aldo Moro Medical School, Bari, Italy
| | - Hermann Einsele
- Department of Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Angelo Vacca
- University of Bari Aldo Moro Medical School, Bari, Italy
| | - Andreas Beilhack
- Department of Medicine II, University Hospital of Würzburg, Würzburg, Germany; 7Interdisciplinary Center for Clinical Research Laboratory, University Hospital of Würzburg, Würzburg, Germany
| |
Collapse
|
16
|
Jia H, Zhang X, Liu X, Qiao R, Liu Y, Lv S, Zhu H, Wang J, Kong Q, Zhang H, Zhang Z. FABP5, a Novel Immune-Related mRNA Prognostic Marker and a Target of Immunotherapy for Multiple Myeloma. Front Med (Lausanne) 2021; 8:667525. [PMID: 34249967 PMCID: PMC8266212 DOI: 10.3389/fmed.2021.667525] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 04/29/2021] [Indexed: 12/31/2022] Open
Abstract
Objective: Multiple myeloma is an incurable hematological malignancy. It is imperative to identify immune markers for early diagnosis and therapy. Here, this study analyzed immune-related mRNAs and assessed their prognostic value and therapeutic potential. Methods: Abnormally expressed immune-related mRNAs were screened between multiple myeloma and normal bone marrow specimens in the GSE47552 and GSE6477 datasets. Their biological functions were then explored. Survival analysis was presented for assessing prognosis-related mRNAs. CIBERSORT was utilized for identifying 22 immune cell compositions of each bone marrow specimen. Correlation between FABP5 mRNA and immune cells was then analyzed in multiple myeloma. Results: Thirty-one immune-related mRNAs were abnormally expressed in multiple myeloma, which were primarily enriched in B cells-related biological processes and pathways. Following validation, FABP5 mRNA was a key risk factor of multiple myeloma. Patients with its up-regulation usually experienced unfavorable outcomes. There were distinct differences in the infiltration levels of B cells naïve, B cells memory, plasma cells, T cells CD4 naïve, resting memory CD4 T cells, activated memory CD4 T cells, Tregs, resting NK cells, M0 macrophages, M1 macrophages, M2 macrophages, and neutrophils between multiple myeloma and normal samples. FABP5 mRNA had correlations to B cells memory, B cells naïve, dendritic cells activated, macrophages M0, macrophages M1, macrophages M2, neutrophils, activated NK cells, resting memory CD4 T cells, CD8 T cells and Tregs. Conclusion: Collectively, our data showed that FABP5 mRNA was related to immune microenvironment, which could be a target of immunotherapy and prognostic marker for multiple myeloma.
Collapse
Affiliation(s)
- Haipeng Jia
- Department of Hematology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Xiaofen Zhang
- Department of Hematology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Xinxin Liu
- Department of Hematology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Ruifang Qiao
- Respiratory Intensive Care Unit, Taian City Central Hospital, Taian, China
| | - Yan Liu
- Department of Hematology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Sulong Lv
- Department of Hematology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Hongbo Zhu
- Department of Hematology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Jie Wang
- Department of Hematology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Qiuhong Kong
- Department of Hematology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Hong Zhang
- Department of Hematology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Zhirong Zhang
- Department of Hematology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| |
Collapse
|
17
|
Li Z, Fu X, Huang J, Zeng P, Huang Y, Chen X, Liang C. Advances in Screening and Development of Therapeutic Aptamers Against Cancer Cells. Front Cell Dev Biol 2021; 9:662791. [PMID: 34095130 PMCID: PMC8170048 DOI: 10.3389/fcell.2021.662791] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/21/2021] [Indexed: 01/10/2023] Open
Abstract
Cancer has become the leading cause of death in recent years. As great advances in medical treatment, emerging therapies of various cancers have been developed. Current treatments include surgery, radiotherapy, chemotherapy, immunotherapy, and targeted therapy. Aptamers are synthetic ssDNA or RNA. They can bind tightly to target molecules due to their unique tertiary structure. It is easy for aptamers to be screened, synthesized, programmed, and chemically modified. Aptamers are emerging targeted drugs that hold great potentials, called therapeutic aptamers. There are few types of therapeutic aptamers that have already been approved by the US Food and Drug Administration (FDA) for disease treatment. Now more and more therapeutic aptamers are in the stage of preclinical research or clinical trials. This review summarized the screening and development of therapeutic aptamers against different types of cancer cells.
Collapse
Affiliation(s)
- Zheng Li
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Xuekun Fu
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Jie Huang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Peiyuan Zeng
- Department of Biochemistry, University of Victoria, Victoria, BC, Canada
| | - Yuhong Huang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Xinxin Chen
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Chao Liang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
18
|
Ito S, Sato T, Maeta T. Role and Therapeutic Targeting of SDF-1α/CXCR4 Axis in Multiple Myeloma. Cancers (Basel) 2021; 13:cancers13081793. [PMID: 33918655 PMCID: PMC8069569 DOI: 10.3390/cancers13081793] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 01/31/2023] Open
Abstract
Simple Summary The SDF-1α/CXCR4 axis plays crucial roles in proliferation, survival, invasion, dissemination, and drug resistance in multiple myeloma. This review summarizes the pleiotropic role of the SDF-1α/CXCR4 axis in multiple myeloma and introduces the SDF-1α/CXCR4 axis-targeted therapies in multiple myeloma. Abstract The C-X-C chemokine receptor type 4 (CXCR4) is a pleiotropic chemokine receptor that is expressed in not only normal hematopoietic cells but also multiple myeloma cells. Its ligand, stromal cell-derived factor 1α (SDF-1α) is produced in the bone marrow microenvironment. The SDF-1α/CXCR4 axis plays a pivotal role in the major physiological processes associated with tumor proliferation, survival, invasion, dissemination, and drug resistance in myeloma cells. This review summarizes the pleiotropic role of the SDF-1α/CXCR4 axis in multiple myeloma and discusses the future perspective in the SDF-1α/CXCR4 axis-targeted therapies in multiple myeloma.
Collapse
|
19
|
Mayo V, Bowles AC, Wubker LE, Ortiz I, Cordoves AM, Cote RJ, Correa D, Agarwal A. Human-derived osteoblast-like cells and pericyte-like cells induce distinct metastatic phenotypes in primary breast cancer cells. Exp Biol Med (Maywood) 2021; 246:971-985. [PMID: 33210551 PMCID: PMC8024509 DOI: 10.1177/1535370220971599] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 10/15/2020] [Indexed: 02/06/2023] Open
Abstract
Approximately 70% of advanced breast cancer patients will develop bone metastases, which accounts for ∼90% of cancer-related mortality. Breast cancer circulating tumor cells (CTCs) establish metastatic tumors in the bone after a close interaction with local bone marrow cells including pericytes and osteoblasts, both related to resident mesenchymal stem/stromal cells (BM-MSCs) progenitors. In vitro recapitulation of the critical cellular players of the bone microenvironment and infiltrating CTCs could provide new insights into their cross-talk during the metastatic cascade, helping in the development of novel therapeutic strategies. Human BM-MSCs were isolated and fractionated according to CD146 presence. CD146+ cells were utilized as pericyte-like cells (PLCs) given the high expression of the marker in perivascular cells, while CD146- cells were induced into an osteogenic phenotype generating osteoblast-like cells (OLCs). Transwell migration assays were performed to establish whether primary breast cancer cells (3384T) were attracted to OLC. Furthermore, proliferation of 3384T breast cancer cells was assessed in the presence of PLC- and OLC-derived conditioned media. Additionally, conditioned media cultures as well as transwell co-cultures of each OLCs and PLCs were performed with 3384T breast cancer cells for gene expression interrogation assessing their induced transcriptional changes with an emphasis on metastatic potential. PLC as well as their conditioned media increased motility and invasion potential of 3384T breast cancer cells, while OLC induced a dormant phenotype, downregulating invasiveness markers related with migration and proliferation. Altogether, these results indicate that PLC distinctively drive 3384T cancer cells to an invasive and migratory phenotype, while OLC induce a quiescence state, thus recapitulating the different phases of the in vivo bone metastatic process. These data show that phenotypic responses from metastasizing cancer cells are influenced by neighboring cells at the bone metastatic niche during the establishment of secondary metastatic tumors.
Collapse
Affiliation(s)
- Vera Mayo
- Department of Biomedical Engineering, DJTMF Biomedical Nanotechnology Institute, University of Miami, Miami, FL 33146, USA
| | - Annie C Bowles
- Department of Biomedical Engineering, DJTMF Biomedical Nanotechnology Institute, University of Miami, Miami, FL 33146, USA
- Department of Orthopedics, UHealth Sports Medicine Institute, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
- Diabetes Research Institute & Cell Transplant Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Laura E Wubker
- Department of Biomedical Engineering, DJTMF Biomedical Nanotechnology Institute, University of Miami, Miami, FL 33146, USA
| | - Ismael Ortiz
- Department of Biomedical Engineering, DJTMF Biomedical Nanotechnology Institute, University of Miami, Miami, FL 33146, USA
| | - Albert M Cordoves
- Department of Biomedical Engineering, DJTMF Biomedical Nanotechnology Institute, University of Miami, Miami, FL 33146, USA
| | - Richard J Cote
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St Louis, MO 63110, USA
| | - Diego Correa
- Department of Orthopedics, UHealth Sports Medicine Institute, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
- Diabetes Research Institute & Cell Transplant Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Ashutosh Agarwal
- Department of Biomedical Engineering, DJTMF Biomedical Nanotechnology Institute, University of Miami, Miami, FL 33146, USA
- Diabetes Research Institute & Cell Transplant Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
20
|
Determination of Potential Therapeutic Targets and Prognostic Markers of Ovarian Cancer by Bioinformatics Analysis. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8883800. [PMID: 33829065 PMCID: PMC8004373 DOI: 10.1155/2021/8883800] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 03/01/2021] [Indexed: 12/11/2022]
Abstract
This study is to study the expression of CXCRs in ovarian cancer tissues and their value in prognosis. The expressions of CXCR1-CXCR7 mRNA between ovarian tumor tissues and normal tissues and in different pathological types of ovarian tumor tissues were compared by ONCOMINE online tool. The relationship between the expression of CXCRs and clinical pathological staging was studied by GEPIA. Kaplan-Meier plotter online tool was used to analyze prognosis. Finally, GO and KEGG analyses and protein interaction network analysis were performed for CXCRs by the DAVID software to predict their function, and cBioPortal was used to identify the key functional genes. The expression of CXCR3/4/7 mRNA in ovarian cancer tissues was higher than that in normal ovarian tissues, and the expression of CXCR4 was the highest (fold change = 306.413, P < 0.05). The expression of CXCR1/2/3/4/7 mRNA in different pathological types of ovarian tumors was significantly different (P < 0.05). Only CXCR5 expression level was associated with tumor staging. Survival analysis showed that high CXCR7 mRNA expression and low CXCR5/6 expression were associated with the shortening of overall survival. High CXCR4/7 expression and low CXCR5/6 expression were associated with the shortening of progression-free survival. High CXCR2/4 expression and low CXCR5/6 expression were closely related to the shortening of postprogressing survival. Protein interaction network analysis showed that GNB1, PTK2, MAPK1, PIK3CA, GNB4, GNA11, KNG1, and ARNT proteins were closely related to the CXC receptor family. CXCR3/4/7 are potential therapeutic targets, and CXCR2/4/5/6/7 are new markers for the prognosis of ovarian cancer.
Collapse
|
21
|
Kuang C, Zhu Y, Guan Y, Xia J, Ouyang J, Liu G, Hao M, Liu J, Guo J, Zhang W, Feng X, Li X, Zhang J, Wu X, Xu H, Li G, Xie L, Fan S, Qiu L, Zhou W. COX2 confers bone marrow stromal cells to promoting TNFα/TNFR1β-mediated myeloma cell growth and adhesion. Cell Oncol (Dordr) 2021; 44:643-659. [PMID: 33646559 DOI: 10.1007/s13402-021-00590-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2021] [Indexed: 12/14/2022] Open
Abstract
PURPOSE Bone marrow stromal cells (BMSCs) have been implicated in multiple myeloma (MM) progression. However, the underlying mechanisms remain largely elusive. Therefore, we aimed to explore key factors in BMSCs that contribute to MM development. METHODS RNA-sequencing was used to perform gene expression profiling in BMSCs. Enzyme-linked immunosorbent assays (ELISAs) were performed to determine the concentrations of PGE2 and TNFα in sera and conditioned media (CM). Western blotting, qRT-PCR and IHC were used to examine the expression of cyclooxygenase 2 (COX2) in BMSCs and to analyze the regulation of TNFα by COX2. Cell growth and adhesion assays were employed to explore the function of COX2 in vitro. A 5T33MMvt-KaLwRij mouse model was used to study the effects of COX2 inhibition in vivo. RESULTS COX2 was found to be upregulated in MM patient-derived BMSCs and to play a critical role in BMSC-induced MM cell proliferation and adhesion. Administration of PGE2 to CM derived from BMSCs promoted MM cell proliferation and adhesion. Conversely, inhibition of COX2 in BMSCs greatly compromised BMSC-induced MM cell proliferation and adhesion. PCR array-based analysis of inflammatory cytokines indicated that COX2 upregulates the expression of TNFα. Subsequent rescue assays showed that an anti-TNFα monoclonal antibody could antagonize COX2-mediated MM cell proliferation and adhesion. Administration of NS398, a specific COX2 inhibitor, inhibited in vivo tumor growth and improved the survival of 5TMM mice. CONCLUSIONS Our results indicate that COX2 contributes to BMSC-induced MM proliferation and adhesion by increasing the secretion of PGE2 and TNFα. Targeting COX2 in BMSCs may serve as a potential therapeutic approach of treating MM.
Collapse
Affiliation(s)
- Chunmei Kuang
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Department of Hematology, Xiangya Hospital; Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, 410078, China
| | - Yinghong Zhu
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Department of Hematology, Xiangya Hospital; Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, 410078, China
| | - Yongjun Guan
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Department of Hematology, Xiangya Hospital; Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, 410078, China
| | - Jiliang Xia
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Department of Hematology, Xiangya Hospital; Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, 410078, China
| | - Jian Ouyang
- Shanghai Center for Bioinformation Technology, Shanghai Academy of Science and Technology, Shanghai, 201203, China
| | - Guizhu Liu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Chinese Academy of Sciences, Shanghai, 200030, China
| | - Mu Hao
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300020, China
| | - Jiabin Liu
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Department of Hematology, Xiangya Hospital; Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, 410078, China
| | - Jiaojiao Guo
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Department of Hematology, Xiangya Hospital; Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, 410078, China
| | - Wenxia Zhang
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300020, China
| | - Xiangling Feng
- Xiang Ya School of Public Health, Central South University, Changsha, 410078, China
| | - Xin Li
- Department of hematology, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Jingyu Zhang
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Department of Hematology, Xiangya Hospital; Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, 410078, China
| | - Xuan Wu
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Department of Hematology, Xiangya Hospital; Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, 410078, China
| | - Hang Xu
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Department of Hematology, Xiangya Hospital; Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, 410078, China
| | - Guancheng Li
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Department of Hematology, Xiangya Hospital; Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, 410078, China
| | - Lu Xie
- Shanghai Center for Bioinformation Technology, Shanghai Academy of Science and Technology, Shanghai, 201203, China
| | - Songqing Fan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Lugui Qiu
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300020, China
| | - Wen Zhou
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Department of Hematology, Xiangya Hospital; Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, 410078, China.
| |
Collapse
|
22
|
Ren Z, Lantermans H, Kuil A, Kraan W, Arenzana-Seisdedos F, Kersten MJ, Spaargaren M, Pals ST. The CXCL12gamma chemokine immobilized by heparan sulfate on stromal niche cells controls adhesion and mediates drug resistance in multiple myeloma. J Hematol Oncol 2021; 14:11. [PMID: 33436043 PMCID: PMC7802348 DOI: 10.1186/s13045-021-01031-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 01/01/2021] [Indexed: 02/06/2023] Open
Abstract
Background The survival and proliferation of multiple myeloma (MM) cells in the bone marrow (BM) critically depend on interaction with stromal cells expressing the chemokine CXCL12. CXCL12 regulates the homing to the BM niche by mediating the transendothelial migration and adhesion/retention of the MM cells. The gamma isoform of CXCL12 (CXCL12γ) has been reported to be highly expressed in mouse BM and to show enhanced biological activity compared to the ‘common’ CXCL12α isoform, mediated by its unique extended C-terminal domain, which binds heparan sulfate proteoglycans (HSPGs) with an extraordinary high affinity.
Here, we investigated the expression of CXCL12γ in human BM and studied its functional role in the interaction of MM cells with BM stromal cells (BMSCs). Methods We assessed CXCL12γ mRNA and protein expression by human BMSCs using qPCR, flow cytometry, and immunohistochemistry. CRISPR-Cas9 was employed to delete CXCL12γ and the heparan sulfate (HS) co-polymerase EXT1 in BMSCs. To study the functional roles of BMSC-derived CXCL12γ and HSPGs in the interaction of MM cells with BMSCs cells, MM cell lines and primary MM cells were co-cultured with BMSCs. Results We observed that CXCL12γ is expressed in situ by reticular stromal cells in both normal and MM BM, as well as by primary BMSC isolates and BMSC lines. Importantly, upon secretion, CXCL12γ, unlike the CXCL12α isoform, was retained on the surface of BMSCs. This membrane retention of CXCL12γ is HSPG mediated, since it was completely annulated by CRISPR-Cas9-mediated deletion of the HS co-polymerase EXT1. CXCL12γ expressed by BMSCs and membrane-retained by HSPGs supported robust adhesion of MM cells to the BMSCs. Specific genetic deletion of either CXCL12γ or EXT1 significantly attenuated the ability of BMSCs to support MM cell adhesion and, in addition, impaired their capacity to protect MM cells from bortezomib-induced cell death. Conclusions We show that CXCL12γ is expressed by human BMSCs and upon secretion is retained on their cell surface by HSPGs. The membrane-bound CXCL12γ controls adhesion of MM cells to the stromal niche and mediates drug resistance. These findings designate CXCL12γ and associated HSPGs as partners in mediating MM–niche interaction and as potential therapeutic targets in MM.
Collapse
Affiliation(s)
- Zemin Ren
- Department of Pathology, Amsterdam University Medical Centers, Loc. AMC, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.,Lymphoma and Myeloma Center Amsterdam - LYMMCARE, and Cancer Center Amsterdam (CCA), Amsterdam, The Netherlands
| | - Hildo Lantermans
- Department of Pathology, Amsterdam University Medical Centers, Loc. AMC, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.,Lymphoma and Myeloma Center Amsterdam - LYMMCARE, and Cancer Center Amsterdam (CCA), Amsterdam, The Netherlands
| | - Annemieke Kuil
- Department of Pathology, Amsterdam University Medical Centers, Loc. AMC, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.,Lymphoma and Myeloma Center Amsterdam - LYMMCARE, and Cancer Center Amsterdam (CCA), Amsterdam, The Netherlands
| | - Willem Kraan
- Department of Pathology, Amsterdam University Medical Centers, Loc. AMC, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.,Lymphoma and Myeloma Center Amsterdam - LYMMCARE, and Cancer Center Amsterdam (CCA), Amsterdam, The Netherlands
| | | | - Marie José Kersten
- Lymphoma and Myeloma Center Amsterdam - LYMMCARE, and Cancer Center Amsterdam (CCA), Amsterdam, The Netherlands.,Department of Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Marcel Spaargaren
- Department of Pathology, Amsterdam University Medical Centers, Loc. AMC, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.,Lymphoma and Myeloma Center Amsterdam - LYMMCARE, and Cancer Center Amsterdam (CCA), Amsterdam, The Netherlands
| | - Steven T Pals
- Department of Pathology, Amsterdam University Medical Centers, Loc. AMC, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands. .,Lymphoma and Myeloma Center Amsterdam - LYMMCARE, and Cancer Center Amsterdam (CCA), Amsterdam, The Netherlands.
| |
Collapse
|
23
|
Signaling Pathway Mediating Myeloma Cell Growth and Survival. Cancers (Basel) 2021; 13:cancers13020216. [PMID: 33435632 PMCID: PMC7827005 DOI: 10.3390/cancers13020216] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary The bone marrow (BM) microenvironment plays a crucial role in pathogenesis of multiple myeloma (MM), and delineation of the intracellular signaling pathways activated in the BM microenvironment in MM cells is essential to develop novel therapeutic strategies to improve patient outcome. Abstract The multiple myeloma (MM) bone marrow (BM) microenvironment consists of different types of accessory cells. Both soluble factors (i.e., cytokines) secreted from these cells and adhesion of MM cells to these cells play crucial roles in activation of intracellular signaling pathways mediating MM cell growth, survival, migration, and drug resistance. Importantly, there is crosstalk between the signaling pathways, increasing the complexity of signal transduction networks in MM cells in the BM microenvironment, highlighting the requirement for combination treatment strategies to blocking multiple signaling pathways.
Collapse
|
24
|
Portella L, Bello AM, Scala S. CXCL12 Signaling in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1302:51-70. [PMID: 34286441 DOI: 10.1007/978-3-030-62658-7_5] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tumor microenvironment (TME) is the local environment of tumor, composed of tumor cells and blood vessels, extracellular matrix (ECM), immune cells, and metabolic and signaling molecules. Chemokines and their receptors play a fundamental role in the crosstalk between tumor cells and TME, regulating tumor-related angiogenesis, specific leukocyte infiltration, and activation of the immune response and directly influencing tumor cell growth, invasion, and cancer progression. The chemokine CXCL12 is a homeostatic chemokine that regulates physiological and pathological process such as inflammation, cell proliferation, and specific migration. CXCL12 activates CXCR4 and CXCR7 chemokine receptors, and the entire axis has been shown to be dysregulated in more than 20 different tumors. CXCL12 binding to CXCR4 triggers multiple signal transduction pathways that regulate intracellular calcium flux, chemotaxis, transcription, and cell survival. CXCR7 binds with high-affinity CXCL12 and with lower-affinity CXCL11, which binds also CXCR3. Although CXCR7 acts as a CXCL12 scavenger through ligand internalization and degradation, it transduces the signal mainly through β-arrestin with a pivotal role in endothelial and neural cells. Recent studies demonstrate that TME rich in CXCL12 leads to resistance to immune checkpoint inhibitors (ICI) therapy and that CXCL12 axis inhibitors sensitize resistant tumors to ICI effect. Thus targeting the CXCL12-mediated axis may control tumor and tumor microenvironment exerting an antitumor dual action. Herein CXCL12 physiology, role in cancer biology and in composite TME, prognostic role, and the relative inhibitors are addressed.
Collapse
Affiliation(s)
- Luigi Portella
- Microenvironment Molecular Targets, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy
| | - Anna Maria Bello
- Microenvironment Molecular Targets, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy
| | - Stefania Scala
- Microenvironment Molecular Targets, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| |
Collapse
|
25
|
Klimienė I, Radzevičius M, Matuzevičienė R, Sinkevič-Belliot K, Kučinskienė ZA, Pečeliūnas V. Adhesion molecule immunophenotype of bone marrow multiple myeloma plasma cells impacts the presence of malignant circulating plasma cells in peripheral blood. Int J Lab Hematol 2020; 43:403-408. [PMID: 33185981 DOI: 10.1111/ijlh.13387] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/28/2020] [Accepted: 10/19/2020] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Multiple myeloma (MM) patients with malignant plasma cells (MMPCs) in their bone marrow (BM) and malignant circulating plasma cells (MMCPCs) in the peripheral blood (PB) are an independent marker of a clinically aggressive disease, and it reflects a poor prognosis defined by a short time to progression and overall survival. We hypothesized that changes in ADM expression on BM MMPCs might contribute to MMCPC presence in the PB of relapsed/refractory multiple myeloma (RRMM) patients. METHODS We assessed the difference in expression of adhesion molecules and receptors related to cell-cell interaction: integrins, hyaluronic acid receptors, chemokine receptors and other proteins on healthy donor PCs, RRMM BM and PB MMPCs. RESULTS Adhesion immunophenotype showed a significant loss of many adhesion molecules when comparing BM MMPCs of MMCPC- and MMCPC+ MM patients (CD49d, CD49e, CD56, CD138). Further decrease of adhesion molecules was shown in MMCPCs (CD49d, CD49e, CD56, CD138, CD58), suggesting that loss of these molecules may allow cells to leave the BM. CONCLUSIONS Loss of adhesion molecule expression enables MMPCs to leave the BM milieu and enter the PB. These changes can be seen in both the PB and BM of MMCPC+ MM patient.
Collapse
Affiliation(s)
- Indrė Klimienė
- Hematology, Oncology and Transfusion Medicine Centre, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Mantas Radzevičius
- Institute of Biomedical Science, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Rėda Matuzevičienė
- Institute of Biomedical Science, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | | | | | - Valdas Pečeliūnas
- Hematology, Oncology and Transfusion Medicine Centre, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| |
Collapse
|
26
|
Zhou X, Dierks A, Kertels O, Kircher M, Schirbel A, Samnick S, Buck AK, Knorz S, Böckle D, Scheller L, Messerschmidt J, Barakat M, Kortüm KM, Rasche L, Einsele H, Lapa C. 18F-FDG, 11C-Methionine, and 68Ga-Pentixafor PET/CT in Patients with Smoldering Multiple Myeloma: Imaging Pattern and Clinical Features. Cancers (Basel) 2020; 12:cancers12082333. [PMID: 32824832 PMCID: PMC7465161 DOI: 10.3390/cancers12082333] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 01/23/2023] Open
Abstract
This study aimed to explore the correlation between imaging patterns and clinical features in patients with smoldering multiple myeloma (SMM) who simultaneously underwent 18F-FDG, 11C-Methionine, and 68Ga-Pentixafor positron emission tomography/computed tomography (PET/CT). We retrieved and analyzed clinical characteristics and PET imaging data of 10 patients with SMM. We found a significant correlation between bone marrow (BM) plasma cell (PC) infiltration and mean standardized uptake values (SUVmean) of lumbar vertebrae L2-L4 on 11C-Methionine PET/CT scans (r = 0.676, p = 0.031) and 68Ga-Pentixafor PET/CT scans (r = 0.839, p = 0.002). However, there was no significant correlation between BM involvement and SUVmean of lumbar vertebrae L2-L4 on 18F-FDG PET/CT scans (r = 0.558, p = 0.093). Similarly, mean target-to-background ratios (TBRmean) of lumbar vertebrae L2-L4 also correlated with bone marrow plasma cell (BMPC) infiltration in 11C-Methionine PET/CT (r = 0.789, p = 0.007) and 68Ga-Pentixafor PET/CT (r = 0.724, p = 0.018) PET/CT. In contrast, we did not observe a significant correlation between BMPC infiltration rate and TBRmean in 18F-FDG PET/CT (r = 0.355, p = 0.313). Additionally, on 11C-Methionine PET/CT scans, we found a significant correlation between BMPC infiltration and TBRmax of lumbar vertebrae L2-L4 (r = 0.642, p = 0.045). In conclusion, 11C-Methionine and 68Ga-Pentixafor PET/CT demonstrate higher sensitivity than 18F-FDG PET/CT in detecting BM involvement in SMM.
Collapse
Affiliation(s)
- Xiang Zhou
- Department of Internal Medicine II, University Hospital of Würzburg, 97080 Würzburg, Germany; (X.Z.); (S.K.); (D.B.); (L.S.); (J.M.); (M.B.); (K.M.K.); (L.R.); (H.E.)
| | - Alexander Dierks
- Department of Nuclear Medicine, University Hospital of Würzburg, 97080 Würzburg, Germany; (A.D.); (M.K.); (A.S.); (S.S.); (A.K.B.)
- Nuclear Medicine, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany
| | - Olivia Kertels
- Department of Diagnostic and Interventional Radiology, University Hospital of Würzburg, 97080 Würzburg, Germany;
| | - Malte Kircher
- Department of Nuclear Medicine, University Hospital of Würzburg, 97080 Würzburg, Germany; (A.D.); (M.K.); (A.S.); (S.S.); (A.K.B.)
- Nuclear Medicine, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany
| | - Andreas Schirbel
- Department of Nuclear Medicine, University Hospital of Würzburg, 97080 Würzburg, Germany; (A.D.); (M.K.); (A.S.); (S.S.); (A.K.B.)
| | - Samuel Samnick
- Department of Nuclear Medicine, University Hospital of Würzburg, 97080 Würzburg, Germany; (A.D.); (M.K.); (A.S.); (S.S.); (A.K.B.)
| | - Andreas K. Buck
- Department of Nuclear Medicine, University Hospital of Würzburg, 97080 Würzburg, Germany; (A.D.); (M.K.); (A.S.); (S.S.); (A.K.B.)
| | - Sebastian Knorz
- Department of Internal Medicine II, University Hospital of Würzburg, 97080 Würzburg, Germany; (X.Z.); (S.K.); (D.B.); (L.S.); (J.M.); (M.B.); (K.M.K.); (L.R.); (H.E.)
| | - David Böckle
- Department of Internal Medicine II, University Hospital of Würzburg, 97080 Würzburg, Germany; (X.Z.); (S.K.); (D.B.); (L.S.); (J.M.); (M.B.); (K.M.K.); (L.R.); (H.E.)
| | - Lukas Scheller
- Department of Internal Medicine II, University Hospital of Würzburg, 97080 Würzburg, Germany; (X.Z.); (S.K.); (D.B.); (L.S.); (J.M.); (M.B.); (K.M.K.); (L.R.); (H.E.)
| | - Janin Messerschmidt
- Department of Internal Medicine II, University Hospital of Würzburg, 97080 Würzburg, Germany; (X.Z.); (S.K.); (D.B.); (L.S.); (J.M.); (M.B.); (K.M.K.); (L.R.); (H.E.)
| | - Mohammad Barakat
- Department of Internal Medicine II, University Hospital of Würzburg, 97080 Würzburg, Germany; (X.Z.); (S.K.); (D.B.); (L.S.); (J.M.); (M.B.); (K.M.K.); (L.R.); (H.E.)
| | - K. Martin Kortüm
- Department of Internal Medicine II, University Hospital of Würzburg, 97080 Würzburg, Germany; (X.Z.); (S.K.); (D.B.); (L.S.); (J.M.); (M.B.); (K.M.K.); (L.R.); (H.E.)
| | - Leo Rasche
- Department of Internal Medicine II, University Hospital of Würzburg, 97080 Würzburg, Germany; (X.Z.); (S.K.); (D.B.); (L.S.); (J.M.); (M.B.); (K.M.K.); (L.R.); (H.E.)
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital of Würzburg, 97080 Würzburg, Germany; (X.Z.); (S.K.); (D.B.); (L.S.); (J.M.); (M.B.); (K.M.K.); (L.R.); (H.E.)
| | - Constantin Lapa
- Department of Nuclear Medicine, University Hospital of Würzburg, 97080 Würzburg, Germany; (A.D.); (M.K.); (A.S.); (S.S.); (A.K.B.)
- Nuclear Medicine, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany
- Correspondence:
| |
Collapse
|
27
|
Liu H, Guo D, Sha Y, Zhang C, Jiang Y, Hong L, Zhang J, Jiang Y, Lu L, Huang H. ANXA7 promotes the cell cycle, proliferation and cell adhesion-mediated drug resistance of multiple myeloma cells by up-regulating CDC5L. Aging (Albany NY) 2020; 12:11100-11115. [PMID: 32526706 PMCID: PMC7346058 DOI: 10.18632/aging.103326] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 04/28/2020] [Indexed: 12/19/2022]
Abstract
This study aimed to investigate whether annexin A7 (ANXA7) could promote the cell cycle, proliferation and cell adhesion-mediated drug resistance (CAM-DR) of multiple myeloma (MM) cells by up-regulating cell division cycle 5-like (CDC5L). As a result, ANXA7 expression was increased in the serum of MM patients and the expression of ANXA7 and CDC5L was also increased in MM cell lines. ANXA7 overexpression promoted the proliferation and cycle of U266 and RPMI8226 cells. The expression of proliferation cell nuclear antigen (PCNA), KI67, cyclin dependent kinase 1 (CDK1) and cyclinB1 in transfected cells was consistent with the changes of proliferation and cell cycle. In co-culture system of BMSC cells and MM cells, expression of CD44, ICAM1 and VCAM1 in MM cells was increased, which was further increased by ANXA7 overexpression. Bortezomib could increase the apoptosis of U266 and RPMI8226 cells. In co-culture system of BMSC cells and MM cells, the promotion effects of bortezomib on apoptosis of MM cells was decreased, which was further suppressed by ANXA7 overexpression. The above effects exerted by ANXA7 overexpression could be reversed by ANXA7 interference. Moreover, ANXA7 was proved to be combined with CDC5L. CDC5L interference could inhibit the promotion effects of ANXA7 overexpression on proliferation and cell cycle and inhibition effects of ANXA7 overexpression on apoptosis of MM cells treated with bortezomib in co-culture system. In conclusion, ANXA7 could promote the cell cycle, proliferation and CAM-DR of MM cells by up-regulating CDC5L.
Collapse
Affiliation(s)
- Haiyan Liu
- Department of Hematology, The Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Dan Guo
- Department of Hematology, The Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Yuou Sha
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chenlu Zhang
- Department of Hematology, The Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Yijing Jiang
- Department of Hematology, The Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Lemin Hong
- Department of Hematology, The Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Jie Zhang
- Department of Hematology, The Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Yuwen Jiang
- Department of Hematology, The Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Ling Lu
- Department of Hematology, The Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Hongming Huang
- Department of Hematology, The Affiliated Hospital of Nantong University, Nantong 226001, China
| |
Collapse
|
28
|
Role of the Bone Marrow Milieu in Multiple Myeloma Progression and Therapeutic Resistance. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20:e752-e768. [PMID: 32651110 DOI: 10.1016/j.clml.2020.05.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/22/2020] [Accepted: 05/29/2020] [Indexed: 01/10/2023]
Abstract
Multiple myeloma (MM) is a cancer of the plasma cells within the bone marrow (BM). Studies have shown that the cellular and noncellular components of the BM milieu, such as cytokines and exosomes, play an integral role in MM pathogenesis and progression by mediating drug resistance and inducing MM proliferation. Moreover, the BM microenvironment of patients with MM facilitates cancer tolerance and immune evasion through the expansion of regulatory immune cells, inhibition of antitumor effector cells, and disruption of the antigen presentation machinery. These are of special relevance, especially in the current era of cancer immunotherapy. An improved understanding of the supportive role of the MM BM microenvironment will allow for the development of future therapies targeting MM in the context of the BM milieu to elicit deeper and more durable responses. In the present review, we have discussed our current understanding of the role of the BM microenvironment in MM progression and resistance to therapy and discuss novel potential approaches to alter its pro-MM function.
Collapse
|
29
|
Mrozik KM, Cheong CM, Hewett DR, Noll JE, Opperman KS, Adwal A, Russell DL, Blaschuk OW, Vandyke K, Zannettino ACW. LCRF-0006, a small molecule mimetic of the N-cadherin antagonist peptide ADH-1, synergistically increases multiple myeloma response to bortezomib. FASEB Bioadv 2020; 2:339-353. [PMID: 32617520 PMCID: PMC7325588 DOI: 10.1096/fba.2019-00073] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 03/31/2020] [Accepted: 04/02/2020] [Indexed: 12/13/2022] Open
Abstract
N-cadherin is a homophilic cell-cell adhesion molecule that plays a critical role in maintaining vascular stability and modulating endothelial barrier permeability. Pre-clinical studies have shown that the N-cadherin antagonist peptide, ADH-1, increases the permeability of tumor-associated vasculature thereby increasing anti-cancer drug delivery to tumors and enhancing tumor response. Small molecule library screens have identified a novel compound, LCRF-0006, that is a mimetic of the classical cadherin His-Ala-Val sequence-containing region of ADH-1. Here, we evaluated the vascular permeability-enhancing and anti-cancer properties of LCRF-0006 using in vitro vascular disruption and cell apoptosis assays, and a well-established pre-clinical model (C57BL/KaLwRij/5TGM1) of the hematological cancer multiple myeloma (MM). We found that LCRF-0006 disrupted endothelial cell junctions in a rapid, transient and reversible manner, and increased vascular permeability in vitro and at sites of MM tumor in vivo. Notably, LCRF-0006 synergistically increased the in vivo anti-MM tumor response to low-dose bortezomib, a frontline anti-MM agent, leading to regression of disease in 100% of mice. Moreover, LCRF-0006 and bortezomib synergistically induced 5TGM1 MM tumor cell apoptosis in vitro. Our findings demonstrate the potential clinical utility of LCRF-0006 to significantly increase bortezomib effectiveness and enhance the depth of tumor response in patients with MM.
Collapse
Affiliation(s)
- Krzysztof M. Mrozik
- Myeloma Research LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesThe University of AdelaideAdelaideAustralia
- Precision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)AdelaideAustralia
| | - Chee M. Cheong
- Myeloma Research LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesThe University of AdelaideAdelaideAustralia
- Precision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)AdelaideAustralia
| | - Duncan R. Hewett
- Myeloma Research LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesThe University of AdelaideAdelaideAustralia
- Precision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)AdelaideAustralia
| | - Jacqueline E. Noll
- Myeloma Research LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesThe University of AdelaideAdelaideAustralia
- Precision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)AdelaideAustralia
| | - Khatora S. Opperman
- Myeloma Research LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesThe University of AdelaideAdelaideAustralia
- Precision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)AdelaideAustralia
| | - Alaknanda Adwal
- Ovarian and Reproductive Cancer Biology LaboratoryRobinson Research InstituteThe University of AdelaideAdelaideAustralia
| | - Darryl L. Russell
- Ovarian and Reproductive Cancer Biology LaboratoryRobinson Research InstituteThe University of AdelaideAdelaideAustralia
| | - Orest W. Blaschuk
- Division of UrologyDepartment of SurgeryMcGill UniversityMontrealCanada
| | - Kate Vandyke
- Myeloma Research LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesThe University of AdelaideAdelaideAustralia
- Precision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)AdelaideAustralia
| | - Andrew C. W. Zannettino
- Myeloma Research LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesThe University of AdelaideAdelaideAustralia
- Precision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)AdelaideAustralia
- Central Adelaide Local Health NetworkAdelaideAustralia
| |
Collapse
|
30
|
New Insights on the Emerging Genomic Landscape of CXCR4 in Cancer: A Lesson from WHIM. Vaccines (Basel) 2020; 8:vaccines8020164. [PMID: 32260318 PMCID: PMC7349554 DOI: 10.3390/vaccines8020164] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 03/30/2020] [Accepted: 03/31/2020] [Indexed: 12/16/2022] Open
Abstract
Deciphering the molecular alterations leading to disease initiation and progression is currently crucial to identify the most relevant targets for precision therapy in cancer patients. Cancers express a complex chemokine network influencing leucocyte infiltration and angiogenesis. Moreover, malignant cells also express a selective repertoire of chemokine receptors that sustain their growth and spread. At present, different cancer types have been shown to overexpress C-X-C chemokine receptor type 4 (CXCR4) and to respond to its ligand C-X-C motif chemokine 12 (CXCL12). The CXCL12/CXCR4 axis influences cancer biology, promoting survival, proliferation, and angiogenesis, and plays a pivotal role in directing migration of cancer cells to sites of metastases, making it a prognostic marker and a therapeutic target. More recently, mutations in the C-terminus of CXCR4 have been identified in the genomic landscape of patients affected by Waldenstrom's macroglobulinemia, a rare B cell neoplasm. These mutations closely resemble those occurring in Warts, Hypogammaglobulinemia, Immunodeficiency, and Myelokathexis (WHIM) syndrome, an immunodeficiency associated with CXCR4 aberrant expression and activity and with chemotherapy resistance in clinical trials. In this review, we summarize the current knowledge on the relevance of CXCR4 mutations in cancer biology, focusing on its importance as predictors of clinical presentation and response to therapy.
Collapse
|
31
|
Crijns H, Vanheule V, Proost P. Targeting Chemokine-Glycosaminoglycan Interactions to Inhibit Inflammation. Front Immunol 2020; 11:483. [PMID: 32296423 PMCID: PMC7138053 DOI: 10.3389/fimmu.2020.00483] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 03/02/2020] [Indexed: 12/12/2022] Open
Abstract
Leukocyte migration into tissues depends on the activity of chemokines that form concentration gradients to guide leukocytes to a specific site. Interaction of chemokines with their specific G protein-coupled receptors (GPCRs) on leukocytes induces leukocyte adhesion to the endothelial cells, followed by extravasation of the leukocytes and subsequent directed migration along the chemotactic gradient. Interaction of chemokines with glycosaminoglycans (GAGs) is crucial for extravasation in vivo. Chemokines need to interact with GAGs on endothelial cells and in the extracellular matrix in tissues in order to be presented on the endothelium of blood vessels and to create a concentration gradient. Local chemokine retention establishes a chemokine gradient and prevents diffusion and degradation. During the last two decades, research aiming at reducing chemokine activity mainly focused on the identification of inhibitors of the interaction between chemokines and their cognate GPCRs. This approach only resulted in limited success. However, an alternative strategy, targeting chemokine-GAG interactions, may be a promising approach to inhibit chemokine activity and inflammation. On this line, proteins derived from viruses and parasites that bind chemokines or GAGs may have the potential to interfere with chemokine-GAG interactions. Alternatively, chemokine mimetics, including truncated chemokines and mutant chemokines, can compete with chemokines for binding to GAGs. Such truncated or mutated chemokines are characterized by a strong binding affinity for GAGs and abrogated binding to their chemokine receptors. Finally, Spiegelmers that mask the GAG-binding site on chemokines, thereby preventing chemokine-GAG interactions, were developed. In this review, the importance of GAGs for chemokine activity in vivo and strategies that could be employed to target chemokine-GAG interactions will be discussed in the context of inflammation.
Collapse
Affiliation(s)
- Helena Crijns
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Vincent Vanheule
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
32
|
Wang H, Gong Y, Liang L, Xiao L, Yi H, Ye M, Roy M, Xia J, Zhou W, Yang C, Shen X, Zhang B, Li Z, Liu J, Zhou H, Xiao X. Lycorine targets multiple myeloma stem cell-like cells by inhibition of Wnt/β-catenin pathway. Br J Haematol 2020; 189:1151-1164. [PMID: 32167591 DOI: 10.1111/bjh.16477] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 12/16/2019] [Indexed: 12/12/2022]
Abstract
Multiple myeloma (MM) is characterised by the proliferation and accumulation of malignant plasma cells in the bone marrow. Despite the progress in treatment over the last few years, MM remains incurable and the majority of patients relapse. MM stem-like cells (MMSCs) have been considered as the main reason for drug resistance and eventual relapse. Currently, therapeutic agents are not enough to eradicate MMSCs, and finding effective strategies to eradicate MMSCs may improve the outcome of patients. Here we showed that lycorine, a natural compound from the Amaryllidaceae species, effectively inhibits the proliferation of myeloma cells from cell lines or patients, mainly through decreasing ALDH1+ cells. Mechanistically, lycorine decreases the MMSC population through inhibition of the Wnt/β-catenin pathway by reducing the β-catenin protein level. Moreover, lycorine could overcome the increasing proportion of ALDH1+ cells caused by bortezomib (BTZ) treatment, and a combination BTZ and lycorine have a synergistic effect on anti-myeloma cells. Furthermore, we found a similar reduction of MMSC characteristics by lycorine in BTZ-resistant MM cells and primary CD138+ plasma cells. Collectively, our findings indicate lycorine as a promising agent to target MMSCs to overcome the drug resistance of BTZ, and that, alone or in combination with BTZ, lycorine is a potential therapeutic strategy for MM treatments.
Collapse
Affiliation(s)
- Haiqin Wang
- Molecular Biology Research Center & Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
| | - Yanfei Gong
- Molecular Biology Research Center & Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
| | - Long Liang
- Molecular Biology Research Center & Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China.,Hematology Department, Xiangya Hospital, Central South University, Changsha, China
| | - Ling Xiao
- Department of Histology and Embryology of School of Basic Medical Sciences, Central South University, Changsha, China
| | - Hui Yi
- Molecular Biology Research Center & Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
| | - Mao Ye
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China
| | - Mridul Roy
- Molecular Biology Research Center & Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China.,Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China
| | - Jiliang Xia
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China
| | - Wen Zhou
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China
| | - Chaoying Yang
- Molecular Biology Research Center & Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
| | - Xiaokai Shen
- Xiangya Medical School, Central South University, Changsha, China
| | - Boxin Zhang
- Xiangya Medical School, Central South University, Changsha, China
| | - Zhenzhen Li
- Molecular Biology Research Center & Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
| | - Jing Liu
- Molecular Biology Research Center & Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
| | - Hui Zhou
- Lymphoma & Hematology Department, the Affiliated Tumor Hospital of Xiangya Medical School of Central South University, Changsha, China
| | - Xiaojuan Xiao
- Molecular Biology Research Center & Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
| |
Collapse
|
33
|
Mouhieddine TH, Ahmad Y, Barlogie B, Jagannath S, Teruya-Feldstein J, Richter J. Increased Muscle CXCR4 Expression in the Setting of Rare Muscle-invasive Multiple Myeloma. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20:e341-e344. [PMID: 32265150 DOI: 10.1016/j.clml.2020.02.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/19/2020] [Accepted: 02/27/2020] [Indexed: 01/02/2023]
Affiliation(s)
- Tarek H Mouhieddine
- Department of Medicine, Icahn School of Medicine at Mount Sinai, The Mount Sinai Hospital, New York, NY.
| | - Yasir Ahmad
- Division of Nuclear Medicine, Department of Radiology, The Mount Sinai Hospital, New York, NY
| | - Bart Barlogie
- Department of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY
| | - Sundar Jagannath
- Department of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY
| | - Julie Teruya-Feldstein
- Department of Pathology, Molecular and Cell-Based Medicine, The Mount Sinai Hospital, New York, NY
| | - Joshua Richter
- Department of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY
| |
Collapse
|
34
|
Natoni A, Farrell ML, Harris S, Falank C, Kirkham-McCarthy L, Macauley MS, Reagan MR, O’Dwyer M. Sialyltransferase inhibition leads to inhibition of tumor cell interactions with E-selectin, VCAM1, and MADCAM1, and improves survival in a human multiple myeloma mouse model. Haematologica 2020; 105:457-467. [PMID: 31101754 PMCID: PMC7012485 DOI: 10.3324/haematol.2018.212266] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 05/16/2019] [Indexed: 12/21/2022] Open
Abstract
Aberrant glycosylation resulting from altered expression of sialyltransferases, such as ST3 β-galactoside α2-3-sialyltransferase 6, plays an important role in disease progression in multiple myeloma (MM). Hypersialylation can lead to increased immune evasion, drug resistance, tumor invasiveness, and disseminated disease. In this study, we explore the in vitro and in vivo effects of global sialyltransferase inhibition on myeloma cells using the pan-sialyltransferase inhibitor 3Fax-Neu5Ac delivered as a per-acetylated methyl ester pro-drug. Specifically, we show in vivo that 3Fax-Neu5Ac improves survival by enhancing bortezomib sensitivity in an aggressive mouse model of MM. However, 3Fax-Neu5Ac treatment of MM cells in vitro did not reverse bortezomib resistance conferred by bone marrow (BM) stromal cells. Instead, 3Fax-Neu5Ac significantly reduced interactions of myeloma cells with E-selectin, MADCAM1 and VCAM1, suggesting that reduced sialylation impairs extravasation and retention of myeloma cells in the BM. Finally, we showed that 3Fax-Neu5Ac alters the post-translational modification of the α4 integrin, which may explain the reduced affinity of α4β1/α4β7 integrins for their counter-receptors. We propose that inhibiting sialylation may represent a valuable strategy to restrict myeloma cells from entering the protective BM microenvironment, a niche in which they are normally protected from chemotherapeutic agents such as bortezomib. Thus, our work demonstrates that targeting sialylation to increase the ratio of circulating to BM-resident MM cells represents a new avenue that could increase the efficacy of other anti-myeloma therapies and holds great promise for future clinical applications.
Collapse
Affiliation(s)
- Alessandro Natoni
- Apoptosis Research Center, National University of Ireland, Galway, Ireland
| | - Mariah L. Farrell
- Maine Medical Center Research Institute, Scarborough, ME, USA,Tufts University School of Medicine, Boston, MA, USA,Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
| | - Sophie Harris
- Maine Medical Center Research Institute, Scarborough, ME, USA,Tufts University School of Medicine, Boston, MA, USA,Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
| | - Carolyne Falank
- Maine Medical Center Research Institute, Scarborough, ME, USA,Tufts University School of Medicine, Boston, MA, USA,Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
| | | | - Matthew S. Macauley
- Department of Chemistry and Department of Medical Microbiology and Immunology, University of Alberta, Alberta, Canada
| | - Michaela R. Reagan
- Maine Medical Center Research Institute, Scarborough, ME, USA,Tufts University School of Medicine, Boston, MA, USA,Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
| | - Michael O’Dwyer
- Apoptosis Research Center, National University of Ireland, Galway, Ireland,Correspondence: MICHAEL O’DWYER,
| |
Collapse
|
35
|
Peled A, Klein S, Beider K, Burger JA, Abraham M. Role of CXCL12 and CXCR4 in the pathogenesis of hematological malignancies. Cytokine 2019; 109:11-16. [PMID: 29903571 DOI: 10.1016/j.cyto.2018.02.020] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 02/18/2018] [Accepted: 02/20/2018] [Indexed: 12/25/2022]
Abstract
The chemokine receptor CXCR4 and its ligand stromal cell-derived factor-1 (SDF-1/CXCL12) are important players in the cross-talk among lymphoma, myeloma and leukemia cells and their microenvironments. In hematological malignancies and solid tumors, the overexpression of CXCR4 on the cell surface has been shown to be responsible for disease progression, increasing tumor cell survival and chemoresistance and metastasis to organs with high CXCL12 levels (e.g., lymph nodes and bone marrow (BM)). Furthermore, the overexpression of CXCR4 has been found to have prognostic significance for disease progression in many type of tumors including lymphoma, leukemia, glioma, and prostate, breast, colorectal, renal, and hepatocellular carcinomas. In leukemia, CXCR4 expression granted leukemic blasts a higher capacity to seed into BM niches, thereby protecting leukemic cells from chemotherapy-induced apoptosis, and was correlated with shorter disease-free survival. In contrast, neutralizing the interaction of CXCL12/CXCR4 with a variety of antagonists induced apoptosis and differentiation and increased the chemosensitivity of lymphoma, myeloma, and leukemia cells. The role of CXCL12 and CXCR4 in the pathogenesis of hematological malignancies and the clinical therapeutic potential of CXCR4 antagonists in these diseases is discussed.
Collapse
MESH Headings
- Apoptosis/immunology
- Cell Survival/physiology
- Chemokine CXCL12/metabolism
- Disease Progression
- Hematologic Neoplasms/drug therapy
- Hematologic Neoplasms/pathology
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Multiple Myeloma/pathology
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
- Prognosis
- Receptors, CXCR4/metabolism
- Tumor Microenvironment/physiology
Collapse
Affiliation(s)
- Amnon Peled
- Goldyne Savad Institute of Gene Therapy, Hebrew University Hospital, P.O.B 12000, Jerusalem 91120, Israel.
| | - Shiri Klein
- Goldyne Savad Institute of Gene Therapy, Hebrew University Hospital, P.O.B 12000, Jerusalem 91120, Israel
| | - Katia Beider
- Hematology Division, Chaim Sheba Medical Center and Tel Aviv University, Tel-Hashomer, Israel
| | - Jan A Burger
- Department of Leukemia, The University of Texas Houston, TX, USA
| | - Michal Abraham
- Goldyne Savad Institute of Gene Therapy, Hebrew University Hospital, P.O.B 12000, Jerusalem 91120, Israel
| |
Collapse
|
36
|
Transcriptional profiling of circulating tumor cells in multiple myeloma: a new model to understand disease dissemination. Leukemia 2019; 34:589-603. [PMID: 31595039 DOI: 10.1038/s41375-019-0588-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/25/2019] [Accepted: 07/23/2019] [Indexed: 12/28/2022]
Abstract
The reason why a few myeloma cells egress from the bone marrow (BM) into peripheral blood (PB) remains unknown. Here, we investigated molecular hallmarks of circulating tumor cells (CTCs) to identify the events leading to myeloma trafficking into the bloodstream. After using next-generation flow to isolate matched CTCs and BM tumor cells from 32 patients, we found high correlation in gene expression at single-cell and bulk levels (r ≥ 0.94, P = 10-16), with only 55 genes differentially expressed between CTCs and BM tumor cells. CTCs overexpressed genes involved in inflammation, hypoxia, or epithelial-mesenchymal transition, whereas genes related with proliferation were downregulated in CTCs. The cancer stem cell marker CD44 was overexpressed in CTCs, and its knockdown significantly reduced migration of MM cells towards SDF1-α and their adhesion to fibronectin. Approximately half (29/55) of genes differentially expressed in CTCs were prognostic in patients with newly-diagnosed myeloma (n = 553; CoMMpass). In a multivariate analysis including the R-ISS, overexpression of CENPF and LGALS1 was significantly associated with inferior survival. Altogether, these results help understanding the presence of CTCs in PB and suggest that hypoxic BM niches together with a pro-inflammatory microenvironment induce an arrest in proliferation, forcing tumor cells to circulate in PB and seek other BM niches to continue growing.
Collapse
|
37
|
Natoni A, Bohara R, Pandit A, O'Dwyer M. Targeted Approaches to Inhibit Sialylation of Multiple Myeloma in the Bone Marrow Microenvironment. Front Bioeng Biotechnol 2019; 7:252. [PMID: 31637237 PMCID: PMC6787837 DOI: 10.3389/fbioe.2019.00252] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 09/17/2019] [Indexed: 11/13/2022] Open
Abstract
Aberrant glycosylation modulates different aspects of tumor biology, and it has long been recognized as a hallmark of cancer. Among the different forms of glycosylation, sialylation, the addition of sialic acid to underlying oligosaccharides, is often dysregulated in cancer. Increased expression of sialylated glycans has been observed in many types of cancer, including multiple myeloma, and often correlates with aggressive metastatic behavior. Myeloma, a cancer of plasma cells, develops in the bone marrow, and colonizes multiple sites of the skeleton including the skull. In myeloma, the bone marrow represents an essential niche where the malignant cells are nurtured by the microenvironment and protected from chemotherapy. Here, we discuss the role of hypersialylation in the metastatic process focusing on multiple myeloma. In particular, we examine how increased sialylation modulates homing of malignant plasma cells into the bone marrow by regulating the activity of molecules important in bone marrow cellular trafficking including selectins and integrins. We also propose that inhibiting sialylation may represent a new therapeutic strategy to overcome bone marrow-mediated chemotherapy resistance and describe different targeted approaches to specifically deliver sialylation inhibitors to the bone marrow microenvironment.
Collapse
Affiliation(s)
- Alessandro Natoni
- Apoptosis Research Centre, School of Medicine, National University of Ireland, Galway, Ireland
| | - Raghvendra Bohara
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Abhay Pandit
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Michael O'Dwyer
- Apoptosis Research Centre, School of Medicine, National University of Ireland, Galway, Ireland
| |
Collapse
|
38
|
Colombo M, Garavelli S, Mazzola M, Platonova N, Giannandrea D, Colella R, Apicella L, Lancellotti M, Lesma E, Ancona S, Palano MT, Barbieri M, Taiana E, Lazzari E, Basile A, Turrini M, Pistocchi A, Neri A, Chiaramonte R. Multiple myeloma exploits Jagged1 and Jagged2 to promote intrinsic and bone marrow-dependent drug resistance. Haematologica 2019; 105:1925-1936. [PMID: 31582544 PMCID: PMC7327642 DOI: 10.3324/haematol.2019.221077] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 09/26/2019] [Indexed: 12/12/2022] Open
Abstract
Multiple myeloma is still incurable due to an intrinsic aggressiveness or, more frequently, to the interactions of malignant plasma cells with the bone marrow (BM) microenvironment. Myeloma cells educate BM cells to support neoplastic cell growth, survival, acquisition of drug resistance resulting in disease relapse. Myeloma microenvironment is characterized by Notch signaling hyperactivation due to the increased expression of Notch1 and 2 and the ligands Jagged1 and 2 in tumor cells. Notch activation influences myeloma cell biology and promotes the reprogramming of BM stromal cells. In this work we demonstrate, in vitro, ex vivo and by using a zebrafish multiple myeloma model, that Jagged inhibition causes a decrease in both myeloma-intrinsic and stromal cell-induced resistance to currently used drugs, i.e. bortezomib, lenalidomide and melphalan. The molecular mechanism of drug resistance involves the chemokine system CXCR4/SDF1α. Myeloma cell-derived Jagged ligands trigger Notch activity in BM stromal cells. These, in turn, secrete higher levels of SDF1α in the BM microenvironment increasing CXCR4 activation in myeloma cells, which is further potentiated by the concomitant increased expression of this receptor induced by Notch activation. Consistently with the augmented pharmacological resistance, SDF1α boosts the expression of BCL2, Survivin and ABCC1. These results indicate that a Jagged-tailored approach may contribute to disrupting the pharmacological resistance due to intrinsic myeloma cell features or to the pathological interplay with BM stromal cells and, conceivably, improve patients' response to standard-of-care therapies.
Collapse
Affiliation(s)
- Michela Colombo
- Department of Health Sciences, Università degli Studi di Milano, Milano
| | - Silvia Garavelli
- Department of Health Sciences, Università degli Studi di Milano, Milano
| | - Mara Mazzola
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milano
| | - Natalia Platonova
- Department of Health Sciences, Università degli Studi di Milano, Milano
| | | | - Raffaella Colella
- Department of Health Sciences, Università degli Studi di Milano, Milano
| | - Luana Apicella
- Department of Health Sciences, Università degli Studi di Milano, Milano
| | | | - Elena Lesma
- Department of Health Sciences, Università degli Studi di Milano, Milano
| | - Silvia Ancona
- Department of Health Sciences, Università degli Studi di Milano, Milano
| | | | - Marzia Barbieri
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milano.,Hematology, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milano
| | - Elisa Taiana
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milano.,Hematology, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milano
| | - Elisa Lazzari
- Department of Health Sciences, Università degli Studi di Milano, Milano
| | - Andrea Basile
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milano
| | - Mauro Turrini
- Department of Hematology, Division of Medicine, Valduce Hospital, Como, Italy
| | - Anna Pistocchi
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milano
| | - Antonino Neri
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milano.,Hematology, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milano
| | | |
Collapse
|
39
|
Wu Y, Zhu X, Shen R, Huang J, Xu X, He S. miR-182 contributes to cell adhesion-mediated drug resistance in multiple myeloma via targeting PDCD4. Pathol Res Pract 2019; 215:152603. [PMID: 31540771 DOI: 10.1016/j.prp.2019.152603] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/01/2019] [Accepted: 08/16/2019] [Indexed: 11/29/2022]
Abstract
miR-182 is a well-described oncogenic miRNA playing a crucial role in the development of many malignancies. However, the role of miR-182 in multiple myeloma (MM) remains unclear. Here, we demonstrate that adhesion of H929 and MM.1S cells to fibronectin could induce miR-182 expression and decrease PDCD4 expression. Furthermore, miR-182 was found to negatively regulate PDCD4 expression in H929 and MM.1S cells. In addition, PDCD4 down-regulation was required for cell adhesion-mediated drug resistance (CAM-DR). Intriguingly, miR-182 up-regulation could promote CAM-DR in H929 and MM.1S cells. Moreover, miR-182 up-regulation and PDCD4 down-regulation enhanced AKT phosphorylation at Ser473 in both H929 and MM.1S cells. Our data suggest that cell adhesion-mediated miR-182 up-regulation and PDCD4 down-regulation may confer drug resistance via enhancing AKT phosphorylation at Ser473.
Collapse
Affiliation(s)
- Yaxun Wu
- Department of Pathology, Affiliated Tumor Hospital of Nantong University, Nantong, 226361, Jiangsu, China
| | - Xinghua Zhu
- Department of Pathology, Affiliated Tumor Hospital of Nantong University, Nantong, 226361, Jiangsu, China
| | - Rong Shen
- Department of Pathology, Affiliated Tumor Hospital of Nantong University, Nantong, 226361, Jiangsu, China
| | - Jieyu Huang
- Department of Pathology, Affiliated Tumor Hospital of Nantong University, Nantong, 226361, Jiangsu, China
| | - Xiaohong Xu
- Department of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong, 226361, Jiangsu, China.
| | - Song He
- Department of Pathology, Affiliated Tumor Hospital of Nantong University, Nantong, 226361, Jiangsu, China.
| |
Collapse
|
40
|
Ullah TR. The role of CXCR4 in multiple myeloma: Cells' journey from bone marrow to beyond. J Bone Oncol 2019; 17:100253. [PMID: 31372333 PMCID: PMC6658931 DOI: 10.1016/j.jbo.2019.100253] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 07/15/2019] [Accepted: 07/16/2019] [Indexed: 12/17/2022] Open
Abstract
CXCR4 is a pleiotropic chemokine receptor which acts through its ligand CXCL12 to regulate diverse physiological processes. CXCR4/CXCL12 axis plays a pivotal role in proliferation, invasion, dissemination and drug resistance in multiple myeloma (MM). Apart from its role in homing, CXCR4 also affects MM cell mobilization and egression out of the bone marrow (BM) which is correlated with distant organ metastasis. Aberrant CXCR4 expression pattern is associated with osteoclastogenesis and tumor growth in MM through its cross talk with various important cell signalling pathways. A deeper insight into understanding of CXCR4 mediated signalling pathways and its role in MM is essential to identify potential therapeutic interventions. The current therapeutic focus is on disrupting the interaction of MM cells with its protective tumor microenvironment where CXCR4 axis plays an essential role. There are still multiple challenges that need to be overcome to target CXCR4 axis more efficiently and to identify novel combination therapies with existing strategies. This review highlights the role of CXCR4 along with its significant interacting partners as a mediator of MM pathogenesis and summarizes the targeted therapies carried out so far.
Collapse
Key Words
- AMC, Angiogenic monomuclear cells
- BM, Bone marrow
- BMSC, Bone marrow stromal cells
- CAM-DR, Cell adhesion‐mediated drug resistance
- CCR–CC, Chemokine receptor
- CCX–CKR, Chemo Centryx–chemokine receptor
- CD4, Cluster of differentiation 4
- CL—CC, Chemokine ligand
- CNS, Central nervous system
- CSCs, Cancer stem cells
- CTAP-III, Connective tissue-activating peptide-III
- CXCL, CXC chemokine ligand
- CXCR, CXC chemokine receptor
- EGF, Epidermal growth factor
- EMD, Extramedullary disease
- EPC, Endothelial progenitor cells
- EPI, Endogenous peptide inhibitor
- ERK, Extracellular signal related kinase
- FGF, Fibroblast growth factor
- G-CSF, Granulocyte colony-stimulating factor
- GPCRs, G protein-coupled chemokine receptors
- HCC, Hepatocellular carcinoma
- HD, Hodgkin's disease
- HGF, Hepatocyte growth factor
- HIF1α, Hypoxia-inducible factor-1 alpha
- HIV, Human Immunodeficiency Virus
- HMGB1, High Mobility Group Box 1
- HPV, Human papillomavirus
- HSC, Hematopoietic stem cells
- IGF, Insulin-like growth factor
- JAK/STAT, Janus Kinase signal transducer and activator of transcription
- JAM-A, Junctional adhesion molecule-A
- JNK, Jun N-terminal kinase
- MAPK, Mitogen Activated Protein Kinase
- MIF, Macrophage migration inhibitory factor
- MM, Multiple myeloma
- MMP, Matrix metalloproteinases
- MRD, Minimal residual disease
- NHL, Non-Hodgkin's lymphoma
- OCL, Octeoclast
- OPG, Osteoprotegerin
- PI3K, phosphoinositide-3 kinase
- PKA, protein kinase A
- PKC, Protein kinase C
- PLC, Phospholipase C
- Pim, Proviral Integrations of Moloney virus
- RANKL, Receptor activator of nuclear factor kappa-Β ligand
- RRMM, Relapsed/refractory multiple myeloma
- SFM-DR, Soluble factor mediated drug resistance
- VEGF, Vascular endothelial growth factor
- VHL, Von Hippel-Lindau
- WHIM, Warts, Hypogammaglobulinemia, Infections, and Myelokathexis
- WM, Waldenström macroglobulinemia
Collapse
|
41
|
Beider K, Bitner H, Voevoda-Dimenshtein V, Rosenberg E, Sirovsky Y, Magen H, Canaani J, Ostrovsky O, Shilo N, Shimoni A, Abraham M, Weiss L, Milyavsky M, Peled A, Nagler A. The mTOR inhibitor everolimus overcomes CXCR4-mediated resistance to histone deacetylase inhibitor panobinostat through inhibition of p21 and mitotic regulators. Biochem Pharmacol 2019; 168:412-428. [PMID: 31325448 DOI: 10.1016/j.bcp.2019.07.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/15/2019] [Indexed: 12/11/2022]
Abstract
Although having promising anti-myeloma properties, the pan-histone deacetylase inhibitor (HDACi) panobinostat lacks therapeutic activity as a single agent. The aim of the current study was to elucidate the mechanisms underlying multiple myeloma (MM) resistance to panobinostat monotherapy and to define strategies to overcome it. Sensitivity of MM cell lines and primary CD138+ cells from MM patients to panobinostat correlated with reduced expression of the chemokine receptor CXCR4, whereas overexpression of CXCR4 in MM cell lines increased their resistance to panobinostat. Decreased sensitivity to HDACi was associated with reversible G0/G1 cell growth arrest while response was characterized by apoptotic cell death. Analysis of intra-cellular signaling mediators revealed the pro-survival mTOR pathway to be regulated by CXCR4 overexpression. Combining panobinostat with mTOR inhibitor everolimus abrogated the resistance to HDACi and induced synergistic cell death. The combination of panobinostat/everolimus resulted in sustained DNA damage and irreversible suppression of proliferation accompanied by robust apoptosis. Gene expression analysis revealed distinct genetic profiles of single versus combined agent exposure. Whereas panobinostat increased the expression of the cell cycle inhibitor p21, co-treatment with everolimus abrogated the increase in p21 and synergistically downregulated the expression of DNA repair genes and mitotic checkpoint regulators. Importantly, the combination of panobinostat with everolimus effectively targeted CXCR4-expressing resistant MM cells in vivo in the BM niche. In summary, our results uncover the mechanism responsible for the strong synergistic anti-MM activity of dual HDAC and mTOR inhibition and provide the rationale for a novel potential therapeutic approach to treat MM.
Collapse
Affiliation(s)
- Katia Beider
- Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, Ramat Gan, Tel Aviv University, Israel
| | - Hanna Bitner
- Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, Ramat Gan, Tel Aviv University, Israel
| | - Valeria Voevoda-Dimenshtein
- Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, Ramat Gan, Tel Aviv University, Israel
| | - Evgenia Rosenberg
- Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, Ramat Gan, Tel Aviv University, Israel
| | - Yaarit Sirovsky
- Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, Ramat Gan, Tel Aviv University, Israel
| | - Hila Magen
- Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, Ramat Gan, Tel Aviv University, Israel
| | - Jonathan Canaani
- Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, Ramat Gan, Tel Aviv University, Israel
| | - Olga Ostrovsky
- Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, Ramat Gan, Tel Aviv University, Israel
| | - Noya Shilo
- Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, Ramat Gan, Tel Aviv University, Israel
| | - Avichai Shimoni
- Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, Ramat Gan, Tel Aviv University, Israel
| | - Michal Abraham
- Goldyne Savad Institute of Gene Therapy, Hebrew University Hospital, Jerusalem, Israel
| | - Lola Weiss
- Goldyne Savad Institute of Gene Therapy, Hebrew University Hospital, Jerusalem, Israel
| | - Michael Milyavsky
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Amnon Peled
- Goldyne Savad Institute of Gene Therapy, Hebrew University Hospital, Jerusalem, Israel
| | - Arnon Nagler
- Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, Ramat Gan, Tel Aviv University, Israel.
| |
Collapse
|
42
|
High-Risk Multiple Myeloma: Integrated Clinical and Omics Approach Dissects the Neoplastic Clone and the Tumor Microenvironment. J Clin Med 2019; 8:jcm8070997. [PMID: 31323969 PMCID: PMC6678140 DOI: 10.3390/jcm8070997] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 06/27/2019] [Accepted: 06/29/2019] [Indexed: 12/11/2022] Open
Abstract
Multiple myeloma (MM) is a genetically heterogeneous disease that includes a subgroup of 10–15% of patients facing dismal survival despite the most intensive treatment. Despite improvements in biological knowledge, MM is still an incurable neoplasia, and therapeutic options able to overcome the relapsing/refractory behavior represent an unmet clinical need. The aim of this review is to provide an integrated clinical and biological overview of high-risk MM, discussing novel therapeutic perspectives, targeting the neoplastic clone and its microenvironment. The dissection of the molecular determinants of the aggressive phenotypes and drug-resistance can foster a better tailored clinical management of the high-risk profile and therapy-refractoriness. Among the current clinical difficulties in MM, patients’ management by manipulating the tumor niche represents a major challenge. The angiogenesis and the stromal infiltrate constitute pivotal mechanisms of a mutual collaboration between MM and the non-tumoral counterpart. Immuno-modulatory and anti-angiogenic therapy hold great efficacy, but variable and unpredictable responses in high-risk MM. The comprehensive understanding of the genetic heterogeneity and MM high-risk ecosystem enforce a systematic bench-to-bedside approach. Here, we provide a broad outlook of novel druggable targets. We also summarize the existing multi-omics-based risk profiling tools, in order to better select candidates for dual immune/vasculogenesis targeting.
Collapse
|
43
|
Becker JH, Gao Y, Soucheray M, Pulido I, Kikuchi E, Rodríguez ML, Gandhi R, Lafuente-Sanchis A, Aupí M, Alcácer Fernández-Coronado J, Martín-Martorell P, Cremades A, Galbis-Caravajal JM, Alcácer J, Christensen CL, Simms P, Hess A, Asahina H, Kahle MP, Al-Shahrour F, Borgia JA, Lahoz A, Insa A, Juan O, Jänne PA, Wong KK, Carretero J, Shimamura T. CXCR7 Reactivates ERK Signaling to Promote Resistance to EGFR Kinase Inhibitors in NSCLC. Cancer Res 2019; 79:4439-4452. [PMID: 31273063 DOI: 10.1158/0008-5472.can-19-0024] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 05/10/2019] [Accepted: 06/27/2019] [Indexed: 12/16/2022]
Abstract
Although EGFR mutant-selective tyrosine kinase inhibitors (TKI) are clinically effective, acquired resistance can occur by reactivating ERK. We show using in vitro models of acquired EGFR TKI resistance with a mesenchymal phenotype that CXCR7, an atypical G protein-coupled receptor, activates the MAPK-ERK pathway via β-arrestin. Depletion of CXCR7 inhibited the MAPK pathway, significantly attenuated EGFR TKI resistance, and resulted in mesenchymal-to-epithelial transition. CXCR7 overexpression was essential in reactivation of ERK1/2 for the generation of EGFR TKI-resistant persister cells. Many patients with non-small cell lung cancer (NSCLC) harboring an EGFR kinase domain mutation, who progressed on EGFR inhibitors, demonstrated increased CXCR7 expression. These data suggest that CXCR7 inhibition could considerably delay and prevent the emergence of acquired EGFR TKI resistance in EGFR-mutant NSCLC. SIGNIFICANCE: Increased expression of the chemokine receptor CXCR7 constitutes a mechanism of resistance to EGFR TKI in patients with non-small cell lung cancer through reactivation of ERK signaling.
Collapse
Affiliation(s)
- Jeffrey H Becker
- Department of Surgery, Division of Cardiothoracic Surgery, University of Illinois at Chicago, Chicago, Illinois.,University of Illinois Hospital & Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, Illinois.,Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Stritch School of Medicine, Maywood, Illinois
| | - Yandi Gao
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Stritch School of Medicine, Maywood, Illinois
| | - Margaret Soucheray
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Stritch School of Medicine, Maywood, Illinois
| | - Ines Pulido
- Departament de Fisiologia, Facultat de Farmacia, Universitat de València, Burjassot, Spain
| | - Eiki Kikuchi
- First department of Medicine, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - María L Rodríguez
- Departament de Fisiologia, Facultat de Farmacia, Universitat de València, Burjassot, Spain
| | - Rutu Gandhi
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Stritch School of Medicine, Maywood, Illinois
| | | | - Miguel Aupí
- Departament de Fisiologia, Facultat de Farmacia, Universitat de València, Burjassot, Spain
| | | | | | - Antonio Cremades
- Department of Pathology, Hospital Universitario de la Ribera, Alzira, Valencia, Spain
| | - José M Galbis-Caravajal
- Department of Thoracic Surgery, Hospital Universitario de la Ribera, Alzira, Valencia, Spain
| | - Javier Alcácer
- Department of Pathology, Hospital Quirónsalud Valencia, Valencia, Spain
| | - Camilla L Christensen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Belfer Institute for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts.,Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Ludwig Center, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Patricia Simms
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Stritch School of Medicine, Maywood, Illinois
| | - Ashley Hess
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Stritch School of Medicine, Maywood, Illinois
| | - Hajime Asahina
- First department of Medicine, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Michael P Kahle
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Stritch School of Medicine, Maywood, Illinois
| | - Fatima Al-Shahrour
- Bioinformatics Unit, Spanish National Cancer Research Centre, Madrid, Spain
| | - Jeffrey A Borgia
- Department of Cell & Molecular Medicine, Rush University Medical Center, Chicago, Illinois
| | - Agustín Lahoz
- Biomarkers and Precision Medicine Unit, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Amelia Insa
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Oscar Juan
- Biomarkers and Precision Medicine Unit, Instituto de Investigación Sanitaria La Fe, Valencia, Spain.,Department of Medical Oncology, Hospital Universitari I Politècnic La Fe, Valencia, Spain
| | - Pasi A Jänne
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Belfer Institute for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts.,Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Bioinformatics Unit, Spanish National Cancer Research Centre, Madrid, Spain
| | - Kwok-Kin Wong
- Laura and Isaac Perlmutter Cancer Center, Division of Hematology and Medical Oncology, New York University, New York, New York
| | - Julian Carretero
- Departament de Fisiologia, Facultat de Farmacia, Universitat de València, Burjassot, Spain.
| | - Takeshi Shimamura
- Department of Surgery, Division of Cardiothoracic Surgery, University of Illinois at Chicago, Chicago, Illinois. .,University of Illinois Hospital & Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, Illinois.,Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Stritch School of Medicine, Maywood, Illinois
| |
Collapse
|
44
|
Mayne ES, Louw S. Good Fences Make Good Neighbors: Human Immunodeficiency Virus and Vascular Disease. Open Forum Infect Dis 2019; 6:ofz303. [PMID: 31737735 PMCID: PMC6847507 DOI: 10.1093/ofid/ofz303] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 06/25/2019] [Indexed: 01/08/2023] Open
Abstract
Cardiovascular disease, venous thrombosis, and microvascular disease in people with HIV (PWH) is predicted to increase in an aging HIV-infected population. Endothelial damage and dysfunction is a risk factor for cardiovascular events in PWH and is characterized by impaired vascular relaxation and decreased nitric oxide availability. Vascular disease has been attributed to direct viral effects, opportunistic infections, chronic inflammation, effects of antiretroviral therapy, and underlying comorbid conditions, like hypertension and use of tobacco. Although biomarkers have been examined to predict and prognosticate thrombotic and cardiovascular disease in this population, more comprehensive validation of risk factors is necessary to ensure patients are managed appropriately. This review examines the pathogenesis of vascular disease in PWH and summarizes the biomarkers used to predict vascular disease in this population.
Collapse
Affiliation(s)
- Elizabeth S Mayne
- Department of Immunology, Faculty of Health Sciences, University of the Witwatersrand and the National Health Laboratory Service
| | - Susan Louw
- Department of Molecular Medicine Faculty of Health Sciences, University of the Witwatersrand and the National Health Laboratory Service, Johannesburg, South Africa
| |
Collapse
|
45
|
Cassis P, Zoja C, Perico L, Remuzzi G. A preclinical overview of emerging therapeutic targets for glomerular diseases. Expert Opin Ther Targets 2019; 23:593-606. [PMID: 31150308 DOI: 10.1080/14728222.2019.1626827] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Animal models have provided significant insights into the mechanisms responsible for the development of glomerular lesions and proteinuria; they have also helped to identify molecules that control the podocyte function as suitable target-specific therapeutics. Areas covered: We discuss putative therapeutic targets for proteinuric glomerular diseases. An exhaustive search for eligible studies was performed in PubMed/MEDLINE. Most of the selected reports were published in the last decade, but we did not exclude older relevant milestone publications. We consider the molecules that regulate podocyte cytoskeletal dynamics and the transcription factors that regulate the expression of slit-diaphragm proteins. There is a focus on SGLT2 and sirtuins which have recently emerged as mediators of podocyte injury and repair. We also examine paracrine signallings involved in the cross-talk of injured podocytes with the neighbouring glomerular endothelial cells and parietal epithelial cells. Expert opinion: There is a need to discover novel therapeutic moleecules with renoprotective effects for those patients with glomerular diseases who do not respond completely to standard therapy. Emerging strategies targeting components of the podocyte cytoskeleton or signallings that regulate cellular communication within the glomerulus are promising avenues for treating glomerular diseases.
Collapse
Affiliation(s)
- Paola Cassis
- a Department of Molecular Medicine , Istituto di Ricerche Farmacologiche Mario Negri IRCCS,Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso , Bergamo , Italy
| | - Carlamaria Zoja
- a Department of Molecular Medicine , Istituto di Ricerche Farmacologiche Mario Negri IRCCS,Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso , Bergamo , Italy
| | - Luca Perico
- a Department of Molecular Medicine , Istituto di Ricerche Farmacologiche Mario Negri IRCCS,Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso , Bergamo , Italy
| | - Giuseppe Remuzzi
- a Department of Molecular Medicine , Istituto di Ricerche Farmacologiche Mario Negri IRCCS,Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso , Bergamo , Italy.,b 'L. Sacco' Department of Biomedical and Clinical Sciences , University of Milan , Milan , Italy
| |
Collapse
|
46
|
Redondo-Muñoz J, García-Pardo A, Teixidó J. Molecular Players in Hematologic Tumor Cell Trafficking. Front Immunol 2019; 10:156. [PMID: 30787933 PMCID: PMC6372527 DOI: 10.3389/fimmu.2019.00156] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 01/17/2019] [Indexed: 12/20/2022] Open
Abstract
The trafficking of neoplastic cells represents a key process that contributes to progression of hematologic malignancies. Diapedesis of neoplastic cells across endothelium and perivascular cells is facilitated by adhesion molecules and chemokines, which act in concert to tightly regulate directional motility. Intravital microscopy provides spatio-temporal views of neoplastic cell trafficking, and is crucial for testing and developing therapies against hematologic cancers. Multiple myeloma (MM), chronic lymphocytic leukemia (CLL), and acute lymphoblastic leukemia (ALL) are hematologic malignancies characterized by continuous neoplastic cell trafficking during disease progression. A common feature of these neoplasias is the homing and infiltration of blood cancer cells into the bone marrow (BM), which favors growth and survival of the malignant cells. MM cells traffic between different BM niches and egress from BM at late disease stages. Besides the BM, CLL cells commonly home to lymph nodes (LNs) and spleen. Likewise, ALL cells also infiltrate extramedullary organs, such as the central nervous system, spleen, liver, and testicles. The α4β1 integrin and the chemokine receptor CXCR4 are key molecules for MM, ALL, and CLL cell trafficking into and out of the BM. In addition, the chemokine receptor CCR7 controls CLL cell homing to LNs, and CXCR4, CCR7, and CXCR3 contribute to ALL cell migration across endothelia and the blood brain barrier. Some of these receptors are used as diagnostic markers for relapse and survival in ALL patients, and their level of expression allows clinicians to choose the appropriate treatments. In CLL, elevated α4β1 expression is an established adverse prognostic marker, reinforcing its role in the disease expansion. Combining current chemotherapies with inhibitors of malignant cell trafficking could represent a useful therapy against these neoplasias. Moreover, immunotherapy using humanized antibodies, CAR-T cells, or immune check-point inhibitors together with agents targeting the migration of tumor cells could also restrict their survival. In this review, we provide a view of the molecular players that regulate the trafficking of neoplastic cells during development and progression of MM, CLL, and ALL, together with current therapies that target the malignant cells.
Collapse
Affiliation(s)
- Javier Redondo-Muñoz
- Department of Immunology, Ophthalmology and ERL, Hospital 12 de Octubre Health Research Institute (imas12), School of Medicine, Complutense University, Madrid, Spain.,Manchester Collaborative Centre for Inflammation Research, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Angeles García-Pardo
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| | - Joaquin Teixidó
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| |
Collapse
|
47
|
Moreno L, Perez C, Zabaleta A, Manrique I, Alignani D, Ajona D, Blanco L, Lasa M, Maiso P, Rodriguez I, Garate S, Jelinek T, Segura V, Moreno C, Merino J, Rodriguez-Otero P, Panizo C, Prosper F, San-Miguel JF, Paiva B. The Mechanism of Action of the Anti-CD38 Monoclonal Antibody Isatuximab in Multiple Myeloma. Clin Cancer Res 2019; 25:3176-3187. [PMID: 30692097 DOI: 10.1158/1078-0432.ccr-18-1597] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 11/26/2018] [Accepted: 01/14/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Knowledge about the mechanism of action (MoA) of monoclonal antibodies (mAb) is required to understand which patients with multiple myeloma (MM) benefit the most from a given mAb, alone or in combination therapy. Although there is considerable research about daratumumab, knowledge about other anti-CD38 mAbs remains scarce. EXPERIMENTAL DESIGN We performed a comprehensive analysis of the MoA of isatuximab. RESULTS Isatuximab induces internalization of CD38 but not its significant release from MM cell surface. In addition, we uncovered an association between levels of CD38 expression and different MoA: (i) Isatuximab was unable to induce direct apoptosis on MM cells with CD38 levels closer to those in patients with MM, (ii) isatuximab sensitized CD38hi MM cells to bortezomib plus dexamethasone in the presence of stroma, (iii) antibody-dependent cellular cytotoxicity (ADCC) was triggered by CD38lo and CD38hi tumor plasma cells (PC), (iv) antibody-dependent cellular phagocytosis (ADCP) was triggered only by CD38hi MM cells, whereas (v) complement-dependent cytotoxicity could be triggered in less than half of the patient samples (those with elevated levels of CD38). Furthermore, we showed that isatuximab depletes CD38hi B-lymphocyte precursors and natural killer (NK) lymphocytes ex vivo-the latter through activation followed by exhaustion and eventually phagocytosis. CONCLUSIONS This study provides a framework to understand response determinants in patients treated with isatuximab based on the number of MoA triggered by CD38 levels of expression, and for the design of effective combinations aimed at capitalizing disrupted tumor-stroma cell protection, augmenting NK lymphocyte-mediated ADCC, or facilitating ADCP in CD38lo MM patients.See related commentary by Malavasi and Faini, p. 2946.
Collapse
Affiliation(s)
- Laura Moreno
- Clinica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Cristina Perez
- Clinica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Aintzane Zabaleta
- Clinica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Irene Manrique
- Clinica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Diego Alignani
- Clinica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Daniel Ajona
- Clinica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369 and CB16/12/00489, Pamplona, Spain.,Solid Tumors Program, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00443, Pamplona, Spain.,Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Laura Blanco
- Clinica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Marta Lasa
- Clinica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Patricia Maiso
- Clinica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Idoia Rodriguez
- Clinica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Sonia Garate
- Clinica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Tomas Jelinek
- Clinica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Victor Segura
- Clinica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Cristina Moreno
- Clinica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Juana Merino
- Clinica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Paula Rodriguez-Otero
- Clinica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Carlos Panizo
- Clinica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Felipe Prosper
- Clinica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Jesus F San-Miguel
- Clinica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Bruno Paiva
- Clinica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369 and CB16/12/00489, Pamplona, Spain.
| |
Collapse
|
48
|
Tang J, Zhu L, Huang Y, Shi B, Zhang S, Gu L, Zhao J, Deng M, Zhu J, Xun H, Wang Y, Wang C. Silencing of LIMD1 promotes proliferation and reverses cell adhesion-mediated drug resistance in non-Hodgkin's lymphoma. Oncol Lett 2019; 17:2993-3000. [PMID: 30854077 DOI: 10.3892/ol.2019.9921] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 11/08/2018] [Indexed: 01/05/2023] Open
Abstract
LIM domains-containing protein 1 (LIMD1) is a tumor suppressor protein downregulated in numerous solid malignancies. However, the functional role of LIMD1 in non-Hodgkin's lymphoma (NHL) remains unclear. In the present study, it was demonstrated that LIMD1 is associated with the proliferation of NHL and cell adhesion mediated-drug resistance (CAM-DR). It was indicated by western blot analysis that LIMD1expression is lower in progressive lymphoma compared with indolent lymphoma. Furthermore, it was indicated that the role of LIMD1 in cell viability and proliferation remains unclear. Finally, the present study demonstrated that LIMD1 serves an important role in CAM-DR by regulating cell cycle progression. Silencing of LIMD1 may reverse CAM-DR in NHL. Therefore, the findings of the present study suggested that LIMD1 may be a potential therapeutic target for patients with NHL.
Collapse
Affiliation(s)
- Jie Tang
- Department of Oncology, Liyang People's Hospital, Liyang, Jiangsu 213300, P.R. China
| | - Liqun Zhu
- Department of Oncology, Liyang People's Hospital, Liyang, Jiangsu 213300, P.R. China
| | - Yuejiao Huang
- Department of Oncology, Affiliated Cancer Hospital of Nantong University, Nantong, Jiangsu 226002, P.R. China
| | - Bing Shi
- Department of Oncology, The Second People's Hospital of Nantong, Nantong, Jiangsu 226002, P.R. China
| | - Shuqing Zhang
- Department of Oncology, The Second People's Hospital of Nantong, Nantong, Jiangsu 226002, P.R. China
| | - Lingli Gu
- Department of Oncology, The Second People's Hospital of Nantong, Nantong, Jiangsu 226002, P.R. China
| | - Jie Zhao
- Department of Pathogenic Biology, Medical College, Nantong University, Nantong, Jiangsu 226002, P.R. China
| | - Minghao Deng
- Department of Pathogenic Biology, Medical College, Nantong University, Nantong, Jiangsu 226002, P.R. China
| | - Jiahao Zhu
- Department of Pathogenic Biology, Medical College, Nantong University, Nantong, Jiangsu 226002, P.R. China
| | - He Xun
- Department of Pathogenic Biology, Medical College, Nantong University, Nantong, Jiangsu 226002, P.R. China
| | - Yuchan Wang
- Department of Pathogenic Biology, Medical College, Nantong University, Nantong, Jiangsu 226002, P.R. China
| | - Chun Wang
- Department of Oncology, Liyang People's Hospital, Liyang, Jiangsu 213300, P.R. China
| |
Collapse
|
49
|
Tau-TCHF Inhibits Spleenic Apoptosis via PI3K-Akt Signaling Pathway in Chickens. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1155:555-563. [PMID: 31468431 DOI: 10.1007/978-981-13-8023-5_51] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Taurine plays an important role in improving immunity, regulating cell proliferation and differentiation, apoptosis and so on. Traditional Chinese herb formula (TCHF) is a wealth of medicine materials for diseases control. There are many studies on Chinese herb formula in inducing cell apoptosis, differentiation and improving animal immunity. The factors in phosphatidylinositol 3-kinase/Protein Kinase (PI3K-Akt) signaling pathway are central regulators of normal cells, which integrates extra-cellular signals into cells and activates affects cell activities including cell proliferation, differentiation and apoptosis. We find the key factors (PIK3CA, PDPK1, AKT1, MDM2, ITGA2B, ITGB1, FAK and p53) in PI3K-Akt signaling pathway by RNA-Seq analysis in our previous research. The overall goal of this study to investigate the influence of taurine TCHF (Tau-TCHF) on cell proliferation, differentiation and apoptosis by estimating the factors above. The layers were fed with normal diet plus 1% of Tau-TCHF and the control group with normal diet to 42 days old. The spleen tissue samples from individual layers were used to analyze the influence of Tau-TCHF on the factors PIK3CA, PDPK1, AKT1, MDM2, ITGA2B, ITGB1, FAK and p53 in PI3K-Akt signaling pathway. The levels of transcription and protein expression of various factors were assessed by quantitative PCR (qPCR) and Western Blot. The results showed that the transcription levels of itgb1, fak, pik3ca, akt1 and mdm2 on 42-day-old chicken spleen tissues were increased significantly in Tau-TCHF group comparing with control group (P < 0.01); the transcription levels of itga2b, pdpk1 and p53 were no significant difference (P > 0.05). The protein levels of PDPK1 and AKT (Ser437) were increased significantly (P < 0.05), but ITGA2B, ITGB1, FAK, PIK3CA, AKT1, MDM2 and p53 had no significant difference (P > 0.05). The results suggest that Tau-TCHF may influence proliferation and differentiation of chickens spleen via regulating PI3K-Akt signaling pathway. And Tau-TCHF may be provided as feed additives in improving the immunity of animals. AKT (Ser473) and PDPK1 may be considered as further targets to study mechanism of Tau-TCHF on anti-apoptosis via PI3K-Akt signaling pathway.
Collapse
|
50
|
Drug resistance in multiple myeloma. Cancer Treat Rev 2018; 70:199-208. [DOI: 10.1016/j.ctrv.2018.09.001] [Citation(s) in RCA: 196] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 08/05/2018] [Accepted: 09/01/2018] [Indexed: 02/07/2023]
|