1
|
Ayoub M, Susin SA, Bauvois B. Tumor Cell Survival Factors and Angiogenesis in Chronic Lymphocytic Leukemia: How Hot Is the Link? Cancers (Basel) 2024; 17:72. [PMID: 39796700 PMCID: PMC11719013 DOI: 10.3390/cancers17010072] [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: 11/12/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Chronic lymphocytic leukemia (CLL) is characterized by the accumulation of neoplastic CD5+/CD19+ B lymphocytes in the blood. These cells migrate to and proliferate in the bone marrow and lymphoid tissues. Despite the development of new therapies for CLL, drug resistance and disease relapse still occur; novel treatment approaches are therefore still needed. Inhibition of the angiogenesis involved in the progression of CLL might be a relevant therapeutic strategy. The literature data indicate that vascular endothelial growth factor, angiopoietin-2, and matrix metalloproteinase-9 are pro-angiogenic factors in CLL. A number of other CLL factors might have pro-angiogenic activity: fibroblast growth factor-2, certain chemokines (such as CXCL-12 and CXCL-2), tumor necrosis factor-α, insulin-like growth factor-1, neutrophil gelatinase-associated lipocalin, and progranulin. All these molecules contribute to the survival, proliferation, and migration of CLL cells. Here, we review the literature on these factors' respective expression profiles and roles in CLL. We also summarize the main results of preclinical and clinical trials of novel agents targeting most of these molecules in a CLL setting. Through the eradication of leukemic cells and the inhibition of angiogenesis, these therapeutic approaches might alter the course of CLL.
Collapse
Affiliation(s)
| | | | - Brigitte Bauvois
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Inserm UMRS 1138, Drug Resistance in Hematological Malignancies Team, F-75006 Paris, France; (M.A.); (S.A.S.)
| |
Collapse
|
2
|
Bauvois B, Nguyen-Khac F, Merle-Béral H, Susin SA. CD38/NAD + glycohydrolase and associated antigens in chronic lymphocytic leukaemia: From interconnected signalling pathways to therapeutic strategies. Biochimie 2024; 227:135-151. [PMID: 39009062 DOI: 10.1016/j.biochi.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 07/12/2024] [Indexed: 07/17/2024]
Abstract
Chronic lymphocytic leukaemia (CLL) is a heterogenous disease characterized by the accumulation of neoplastic CD5+/CD19+ B lymphocytes. The spreading of the leukaemia relies on the CLL cell's ability to survive in the blood and migrate to and proliferate within the bone marrow and lymphoid tissues. Some patients with CLL are either refractory to the currently available therapies or relapse after treatment; this emphasizes the need for novel therapeutic strategies that improving clinical responses and overcome drug resistance. CD38 is a marker of a poor prognosis and governs a set of survival, proliferation and migration signals that contribute to the pathophysiology of CLL. The literature data evidence a spatiotemporal association between the cell surface expression of CD38 and that of other CLL antigens, such as the B-cell receptor (BCR), CD19, CD26, CD44, the integrin very late antigen 4 (VLA4), the chemokine receptor CXCR4, the vascular endothelial growth factor receptor-2 (VEGF-R2), and the neutrophil gelatinase-associated lipocalin receptor (NGAL-R). Most of these proteins contribute to CLL cell survival, proliferation and trafficking, and cooperate with CD38 in multilayered signal transduction processes. In general, these antigens have already been validated as therapeutic targets in cancer, and a broad repertoire of specific monoclonal antibodies and derivatives are available. Here, we review the state of the art in this field and examine the therapeutic opportunities for cotargeting CD38 and its partners in CLL, e.g. by designing novel bi-/trispecific antibodies.
Collapse
MESH Headings
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- ADP-ribosyl Cyclase 1/metabolism
- ADP-ribosyl Cyclase 1/immunology
- Signal Transduction
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Membrane Glycoproteins/metabolism
- Membrane Glycoproteins/immunology
- Animals
Collapse
Affiliation(s)
- Brigitte Bauvois
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Inserm UMRS1138, Drug Resistance in Hematological Malignancies Team, F-75006, Paris, France.
| | - Florence Nguyen-Khac
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Inserm UMRS1138, Drug Resistance in Hematological Malignancies Team, F-75006, Paris, France; Sorbonne Université, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Service d'Hématologie Biologique, F-75013, Paris, France.
| | - Hélène Merle-Béral
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Inserm UMRS1138, Drug Resistance in Hematological Malignancies Team, F-75006, Paris, France.
| | - Santos A Susin
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Inserm UMRS1138, Drug Resistance in Hematological Malignancies Team, F-75006, Paris, France.
| |
Collapse
|
3
|
Eroz I, Kakkar PK, Lazar RA, El-Jawhari J. Mesenchymal Stem Cells in Myelodysplastic Syndromes and Leukaemia. Biomedicines 2024; 12:1677. [PMID: 39200142 PMCID: PMC11351218 DOI: 10.3390/biomedicines12081677] [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: 06/26/2024] [Revised: 07/17/2024] [Accepted: 07/24/2024] [Indexed: 09/01/2024] Open
Abstract
Mesenchymal stem cells (MSCs) are one of the main residents in the bone marrow (BM) and have an essential role in the regulation of haematopoietic stem cell (HSC) differentiation and proliferation. Myelodysplastic syndromes (MDSs) are a group of myeloid disorders impacting haematopoietic stem and progenitor cells (HSCPs) that are characterised by BM failure, ineffective haematopoiesis, cytopenia, and a high risk of transformation through the expansion of MDS clones together with additional genetic defects. It has been indicated that MSCs play anti-tumorigenic roles such as in cell cycle arrest and pro-tumorigenic roles including the induction of metastasis in MDS and leukaemia. Growing evidence has shown that MSCs have impaired functions in MDS, such as decreased proliferation capacity, differentiation ability, haematopoiesis support, and immunomodulation function and increased inflammatory alterations within the BM through some intracellular pathways such as Notch and Wnt and extracellular modulators abnormally secreted by MSCs, including increased expression of inflammatory factors and decreased expression of haematopoietic factors, contributing to the development and progression of MDSs. Therefore, MSCs can be targeted for the treatment of MDSs and leukaemia. However, it remains unclear what drives MSCs to behave abnormally. In this review, dysregulations in MSCs and their contributions to myeloid haematological malignancies will be discussed.
Collapse
Affiliation(s)
- Ilayda Eroz
- Biosciences Department, School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, UK (P.K.K.); (R.A.L.)
| | - Prabneet Kaur Kakkar
- Biosciences Department, School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, UK (P.K.K.); (R.A.L.)
| | - Renal Antoinette Lazar
- Biosciences Department, School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, UK (P.K.K.); (R.A.L.)
| | - Jehan El-Jawhari
- Biosciences Department, School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, UK (P.K.K.); (R.A.L.)
- Clinical Pathology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
4
|
Piergentili R, Sechi S. Non-Coding RNAs of Mitochondrial Origin: Roles in Cell Division and Implications in Cancer. Int J Mol Sci 2024; 25:7498. [PMID: 39000605 PMCID: PMC11242419 DOI: 10.3390/ijms25137498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024] Open
Abstract
Non-coding RNAs (ncRNAs) are a heterogeneous group, in terms of structure and sequence length, consisting of RNA molecules that do not code for proteins. These ncRNAs have a central role in the regulation of gene expression and are virtually involved in every process analyzed, ensuring cellular homeostasis. Although, over the years, much research has focused on the characterization of non-coding transcripts of nuclear origin, improved bioinformatic tools and next-generation sequencing (NGS) platforms have allowed the identification of hundreds of ncRNAs transcribed from the mitochondrial genome (mt-ncRNA), including long non-coding RNA (lncRNA), circular RNA (circRNA), and microRNA (miR). Mt-ncRNAs have been described in diverse cellular processes such as mitochondrial proteome homeostasis and retrograde signaling; however, the function of the majority of mt-ncRNAs remains unknown. This review focuses on a subgroup of human mt-ncRNAs whose dysfunction is associated with both failures in cell cycle regulation, leading to defects in cell growth, cell proliferation, and apoptosis, and the development of tumor hallmarks, such as cell migration and metastasis formation, thus contributing to carcinogenesis and tumor development. Here we provide an overview of the mt-ncRNAs/cancer relationship that could help the future development of new biomedical applications in the field of oncology.
Collapse
Affiliation(s)
| | - Stefano Sechi
- Istituto di Biologia e Patologia Molecolari del Consiglio Nazionale delle Ricerche, Dipartimento di Biologia e Biotecnologie, Università Sapienza di Roma, Piazzale Aldo Moro 5, 00185 Rome, Italy;
| |
Collapse
|
5
|
Vitale C, Griggio V, Todaro M, Riganti C, Jones R, Boccellato E, Perutelli F, Arruga F, Vaisitti T, Efremov DG, Deaglio S, Landesman Y, Bruno B, Coscia M. Anti-tumor activity of selinexor in combination with antineoplastic agents in chronic lymphocytic leukemia. Sci Rep 2023; 13:16950. [PMID: 37805613 PMCID: PMC10560255 DOI: 10.1038/s41598-023-44039-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 10/03/2023] [Indexed: 10/09/2023] Open
Abstract
Despite recent relevant therapeutic progresses, chronic lymphocytic leukemia (CLL) remains an incurable disease. Selinexor, an oral inhibitor of the nuclear export protein XPO1, is active as single agent in different hematologic malignancies, including CLL. The purpose of this study was to evaluate the anti-tumor effects of selinexor, used in combination with chemotherapy drugs (i.e. fludarabine and bendamustine) or with the PI3Kδ inhibitor idelalisib in CLL. Our results showed a significant decrease in CLL cell viability after treatment with selinexor-containing drug combinations compared to each single compound, with demonstration of synergistic cytotoxic effects. Interestingly, this drug synergism was exerted also in the presence of the protective effect of stromal cells. From the molecular standpoint, the synergistic cytotoxic activity of selinexor plus idelalisib was associated with increased regulatory effects of this drug combination on the tumor suppressors FOXO3A and IkBα compared to each single compound. Finally, selinexor was also effective in potentiating the in vivo anti-tumor effects of the PI3Kδ inhibitor in mice treated with the drug combination compared to single agents. Our data provide preclinical evidence of the synergism and potential efficacy of a combination treatment targeting XPO1 and PI3Kδ in CLL.
Collapse
Affiliation(s)
- Candida Vitale
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, 10126, Turin, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126, Turin, Italy
| | - Valentina Griggio
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, 10126, Turin, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126, Turin, Italy
| | - Maria Todaro
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, 10126, Turin, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126, Turin, Italy
| | - Chiara Riganti
- Department of Oncology, University of Torino, 10126, Turin, Italy
| | - Rebecca Jones
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, 10126, Turin, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126, Turin, Italy
| | - Elia Boccellato
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, 10126, Turin, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126, Turin, Italy
| | - Francesca Perutelli
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, 10126, Turin, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126, Turin, Italy
| | - Francesca Arruga
- Department of Medical Sciences, University of Torino, 10126, Turin, Italy
| | - Tiziana Vaisitti
- Department of Medical Sciences, University of Torino, 10126, Turin, Italy
| | - Dimitar G Efremov
- Molecular Hematology, International Centre for Genetic Engineering and Biotechnology, 34149, Trieste, Italy
| | - Silvia Deaglio
- Department of Medical Sciences, University of Torino, 10126, Turin, Italy
| | | | - Benedetto Bruno
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, 10126, Turin, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126, Turin, Italy
| | - Marta Coscia
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, 10126, Turin, Italy.
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126, Turin, Italy.
| |
Collapse
|
6
|
Koehrer S, Burger JA. Chronic Lymphocytic Leukemia: Disease Biology. Acta Haematol 2023; 147:8-21. [PMID: 37717577 PMCID: PMC11753505 DOI: 10.1159/000533610] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 08/13/2023] [Indexed: 09/19/2023]
Abstract
BACKGROUND B-cell receptor (BCR) signaling is crucial for normal B-cell development and adaptive immunity. In chronic lymphocytic leukemia (CLL), the malignant B cells display many features of normal mature B lymphocytes, including the expression of functional B-cell receptors (BCRs). Cross talk between CLL cells and the microenvironment in secondary lymphatic organs results in BCR signaling and BCR-driven proliferation of the CLL cells. This critical pathomechanism can be targeted by blocking BCR-related kinases (BTK, PI3K, spleen tyrosine kinase) using small-molecule inhibitors. Among these targets, Bruton tyrosine kinase (BTK) inhibitors have the highest therapeutic efficacy; they effectively block leukemia cell proliferation and generally induce durable remissions in CLL patients, even in patients with high-risk disease. By disrupting tissue homing receptor (i.e., chemokine receptor and adhesion molecule) signaling, these kinase inhibitors also mobilize CLL cells from the lymphatic tissues into the peripheral blood (PB), causing a transient redistribution lymphocytosis, thereby depriving CLL cells from nurturing factors within the tissue niches. SUMMARY The clinical success of the BTK inhibitors in CLL underscores the central importance of the BCR in CLL pathogenesis. Here, we review CLL pathogenesis with a focus on the role of the BCR and other microenvironment cues. KEY MESSAGES (i) CLL cells rely on signals from their microenvironment for proliferation and survival. (ii) These signals are mediated by the BCR as well as chemokine and integrin receptors and their respective ligands. (iii) Targeting the CLL/microenvironment interaction with small-molecule inhibitors provides a highly effective treatment strategy, even in high-risk patients.
Collapse
Affiliation(s)
- Stefan Koehrer
- Department of Laboratory Medicine, Klinik Donaustadt, Vienna, Austria
- Labdia Labordiagnostik, Clinical Genetics, Vienna, Austria
- St. Anna Children’s Cancer Research Institute (CCRI), Vienna, Austria
| | - Jan A. Burger
- Department of Leukemia, The University of Texas, M.D. Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
7
|
Kaufman M, Yan XJ, Li W, Ghia EM, Langerak AW, Rassenti LZ, Belessi C, Kay NE, Davi F, Byrd JC, Pospisilova S, Brown JR, Catherwood M, Davis Z, Oscier D, Montillo M, Trentin L, Rosenquist R, Ghia P, Barrientos JC, Kolitz JE, Allen SL, Rai KR, Stamatopoulos K, Kipps TJ, Neuberg D, Chiorazzi N. Impact of the Types and Relative Quantities of IGHV Gene Mutations in Predicting Prognosis of Patients With Chronic Lymphocytic Leukemia. Front Oncol 2022; 12:897280. [PMID: 35903706 PMCID: PMC9315922 DOI: 10.3389/fonc.2022.897280] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Patients with CLL with mutated IGHV genes (M-CLL) have better outcomes than patients with unmutated IGHVs (U-CLL). Since U-CLL usually express immunoglobulins (IGs) that are more autoreactive and more effectively transduce signals to leukemic B cells, B-cell receptor (BCR) signaling is likely at the heart of the worse outcomes of CLL cases without/few IGHV mutations. A corollary of this conclusion is that M-CLL follow less aggressive clinical courses because somatic IGHV mutations have altered BCR structures and no longer bind stimulatory (auto)antigens and so cannot deliver trophic signals to leukemic B cells. However, the latter assumption has not been confirmed in a large patient cohort. We tried to address the latter by measuring the relative numbers of replacement (R) mutations that lead to non-conservative amino acid changes (Rnc) to the combined numbers of conservative (Rc) and silent (S) amino acid R mutations that likely do not or cannot change amino acids, "(S+Rc) to Rnc IGHV mutation ratio". When comparing time-to-first-treatment (TTFT) of patients with (S+Rc)/Rnc ≤ 1 and >1, TTFTs were similar, even after matching groups for equal numbers of samples and identical numbers of mutations per sample. Thus, BCR structural change might not be the main reason for better outcomes for M-CLL. Since the total number of IGHV mutations associated better with longer TTFT, better clinical courses appear due to the biologic state of a B cell having undergone many stimulatory events leading to IGHV mutations. Analyses of larger patient cohorts will be needed to definitively answer this question.
Collapse
Affiliation(s)
- Matthew Kaufman
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Xiao-Jie Yan
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Wentian Li
- The Robert S. Boas Center for Genomics & Human Genetics, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Emanuela M. Ghia
- Center for Novel Therapeutics, Moores Cancer Center, University of California, San Diego, La Jolla, CA, United States
| | - Anton W. Langerak
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Laura Z. Rassenti
- Center for Novel Therapeutics, Moores Cancer Center, University of California, San Diego, La Jolla, CA, United States
| | | | - Neil E. Kay
- Division of Hematology, Mayo Clinic, Rochester, MN, United States
| | - Frederic Davi
- Department of Biological Hematology, Hôpital Pitié-Salpêtrière (AP-HP), Sorbonne Université, Paris, France
| | - John C. Byrd
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Sarka Pospisilova
- Department of Internal Medicine - Hematology and Oncology and Department of Medical Genetics and Genomics, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Jennifer R. Brown
- Chronic Lymphocytic Leukemia Center, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Mark Catherwood
- Clinical Hematology, Belfast City Hospital, Belfast, Ireland
| | - Zadie Davis
- Department of Molecular Pathology, Royal Bournemouth Hospital, Bournemouth, United Kingdom
| | - David Oscier
- Department of Hematology, Royal Bournemouth Hospital, Bournemouth, United Kingdom
| | - Marco Montillo
- Department of Hematology & Oncology, Niguarda Cancer Center, Niguarda Hospital, Milan, Italy
| | - Livio Trentin
- Hematology Unit, Department of Medicine-(DIMED), University of Padua University Hospital, Padua, Italy
| | - Richard Rosenquist
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Paolo Ghia
- Division of Experimental Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Jacqueline C. Barrientos
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Uniondale, NY, United States
- Departments of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Uniondale, NY, United States
- Northwell Health Cancer Institute, Lake Success, NY, United States
| | - Jonathan E. Kolitz
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- Northwell Health Cancer Institute, Lake Success, NY, United States
| | - Steven L. Allen
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- Northwell Health Cancer Institute, Lake Success, NY, United States
| | - Kanti R. Rai
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Uniondale, NY, United States
- Departments of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Uniondale, NY, United States
- Northwell Health Cancer Institute, Lake Success, NY, United States
| | - Kostas Stamatopoulos
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece
| | - Thomas J. Kipps
- Center for Novel Therapeutics, Moores Cancer Center, University of California, San Diego, La Jolla, CA, United States
| | - Donna Neuberg
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Nicholas Chiorazzi
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Uniondale, NY, United States
- Departments of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Uniondale, NY, United States
- Northwell Health Cancer Institute, Lake Success, NY, United States
| |
Collapse
|
8
|
Ruiz-Aparicio PF, Vernot JP. Bone Marrow Aging and the Leukaemia-Induced Senescence of Mesenchymal Stem/Stromal Cells: Exploring Similarities. J Pers Med 2022; 12:jpm12050716. [PMID: 35629139 PMCID: PMC9147878 DOI: 10.3390/jpm12050716] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/20/2022] [Accepted: 04/27/2022] [Indexed: 12/17/2022] Open
Abstract
Bone marrow aging is associated with multiple cellular dysfunctions, including perturbed haematopoiesis, the propensity to haematological transformation, and the maintenance of leukaemia. It has been shown that instructive signals from different leukemic cells are delivered to stromal cells to remodel the bone marrow into a supportive leukemic niche. In particular, cellular senescence, a physiological program with both beneficial and deleterious effects on the health of the organisms, may be responsible for the increased incidence of haematological malignancies in the elderly and for the survival of diverse leukemic cells. Here, we will review the connection between BM aging and cellular senescence and the role that these processes play in leukaemia progression. Specifically, we discuss the role of mesenchymal stem cells as a central component of the supportive niche. Due to the specificity of the genetic defects present in leukaemia, one would think that bone marrow alterations would also have particular changes, making it difficult to envisage a shared therapeutic use. We have tried to summarize the coincident features present in BM stromal cells during aging and senescence and in two different leukaemias, acute myeloid leukaemia, with high frequency in the elderly, and B-acute lymphoblastic leukaemia, mainly a childhood disease. We propose that mesenchymal stem cells are similarly affected in these different leukaemias, and that the changes that we observed in terms of cellular function, redox balance, genetics and epigenetics, soluble factor repertoire and stemness are equivalent to those occurring during BM aging and cellular senescence. These coincident features may be used to explore strategies useful to treat various haematological malignancies.
Collapse
Affiliation(s)
- Paola Fernanda Ruiz-Aparicio
- Grupo de Investigación Fisiología Celular y Molecular, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá 111321, Colombia;
| | - Jean-Paul Vernot
- Grupo de Investigación Fisiología Celular y Molecular, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá 111321, Colombia;
- Instituto de Investigaciones Biomédicas, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá 111321, Colombia
- Correspondence:
| |
Collapse
|
9
|
Sensitization to Drug Treatment in Precursor B-Cell Acute Lymphoblastic Leukemia Is Not Achieved by Stromal NF-κB Inhibition of Cell Adhesion but by Stromal PKC-Dependent Inhibition of ABC Transporters Activity. Molecules 2021; 26:molecules26175366. [PMID: 34500796 PMCID: PMC8433757 DOI: 10.3390/molecules26175366] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/26/2021] [Accepted: 08/31/2021] [Indexed: 01/10/2023] Open
Abstract
Cell adhesion to stromal support and the associated intracellular signaling are central to drug resistance, therefore blocking both has been effective in increasing drug sensitization in leukemia. The stromal Ser/Thr protein kinase C (PKC) has been found to be important for conferring protection to leukemic cells. We aimed at elucidating the intracellular signals connected to cell adhesion and to stromal PKC. We found that NF-κB and Akt were up-regulated in mesenchymal stem cells (MSC) after binding of B-cell acute lymphoblastic leukemia (B-ALL) cells. Nevertheless, Akt inhibition did not induce B-ALL cell detachment. In spite of a clear activation of the NF-κB signaling pathway after B-ALL cell binding (up-regulation NF-κB1/2, and down-regulation of the IKBε and IKBα inhibitors) and an important reduction in cell adhesion after NF-κB inhibition, sensitization to the drug treatment was not observed. This was opposite to the PKC inhibitors Enzastaurin and HKPS, a novel chimeric peptide inhibitor, that were able to increase sensitization to dexamethasone, methotrexate, and vincristine. PLCγ1, Erk1/2, and CREB appear to be related to PKC signaling and PKC effect on drug sensitization since they were contra-regulated by HKPS when compared to dexamethasone-treated cells. Additionally, PKC inhibition by HKPS, but not by Enzastaurin, in MSC reduced the activity of three ABC transporters in leukemic cells treated with dexamethasone, a new indirect mechanism to increase sensitization to drug treatment in B-ALL cells. Our results show the validity of targeting the functional characteristic acquired and modulated during cell-to-cell interactions occurring in the leukemic niche.
Collapse
|
10
|
Vitale C, Griggio V, Riganti C, Todaro M, Kopecka J, Jones R, Salvetti C, Boccellato E, Perutelli F, Voena C, Godio L, Boccadoro M, Coscia M. Targeting HIF-1α Regulatory Pathways as a Strategy to Hamper Tumor-Microenvironment Interactions in CLL. Cancers (Basel) 2021; 13:cancers13122883. [PMID: 34207596 PMCID: PMC8229189 DOI: 10.3390/cancers13122883] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/26/2021] [Accepted: 06/04/2021] [Indexed: 12/25/2022] Open
Abstract
The hypoxia-inducible factor 1 (HIF-1) and the CXCL12/CXCR4 axis regulate the interaction of chronic lymphocytic leukemia cells and the tumor microenvironment. However, the interconnections occurring between HIF-1 and the CXCL12/CXCR4 axis are not fully elucidated. Here, we demonstrate that the CXCL12/CXCR4 axis plays a pivotal role in the positive regulation of the α subunit of HIF-1 (HIF-1α) that occurs in CLL cells co-cultured with stromal cells (SC). Inhibitors acting at different levels on CXCR4 downstream signalling counteract the SC-induced HIF-1α upregulation in CLL cells, also hindering the SC-mediated pro-survival effect. HIF-1α inhibition also exerts off-tumor effects on the SC component, inducing the downregulation of target genes, including CXCL12. Consistently, our data show that pretreatment of leukemic cells and/or SC with idelalisib effectively abrogates the SC-mediated survival support. A combined on-tumor and off-tumor inhibition of HIF-1α was also observed in idelalisib-treated patients, who showed, along with a downregulation of HIF-1α target genes in leukemic cells, a significant decrease in CXCL12 serum concentration and changes in the bone marrow microenvironment. Our data demonstrate that the targeting of HIF-1α or its regulatory pathways acts at the tumor- and SC-level, and may be an appealing strategy to overcome the microenvironment-mediated protection of CLL cells.
Collapse
Affiliation(s)
- Candida Vitale
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, via Genova 3, 10126 Torino, Italy; (C.V.); (V.G.); (M.T.); (R.J.); (C.S.); (E.B.); (F.P.); (M.B.)
- Department of Molecular Biotechnology and Health Sciences, University of Torino, via Nizza 52, 10126 Torino, Italy;
| | - Valentina Griggio
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, via Genova 3, 10126 Torino, Italy; (C.V.); (V.G.); (M.T.); (R.J.); (C.S.); (E.B.); (F.P.); (M.B.)
- Department of Molecular Biotechnology and Health Sciences, University of Torino, via Nizza 52, 10126 Torino, Italy;
| | - Chiara Riganti
- Department of Oncology, University of Torino, via Santena 5, 10126 Torino, Italy; (C.R.); (J.K.)
| | - Maria Todaro
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, via Genova 3, 10126 Torino, Italy; (C.V.); (V.G.); (M.T.); (R.J.); (C.S.); (E.B.); (F.P.); (M.B.)
- Department of Molecular Biotechnology and Health Sciences, University of Torino, via Nizza 52, 10126 Torino, Italy;
| | - Joanna Kopecka
- Department of Oncology, University of Torino, via Santena 5, 10126 Torino, Italy; (C.R.); (J.K.)
| | - Rebecca Jones
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, via Genova 3, 10126 Torino, Italy; (C.V.); (V.G.); (M.T.); (R.J.); (C.S.); (E.B.); (F.P.); (M.B.)
- Department of Molecular Biotechnology and Health Sciences, University of Torino, via Nizza 52, 10126 Torino, Italy;
| | - Chiara Salvetti
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, via Genova 3, 10126 Torino, Italy; (C.V.); (V.G.); (M.T.); (R.J.); (C.S.); (E.B.); (F.P.); (M.B.)
- Department of Molecular Biotechnology and Health Sciences, University of Torino, via Nizza 52, 10126 Torino, Italy;
| | - Elia Boccellato
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, via Genova 3, 10126 Torino, Italy; (C.V.); (V.G.); (M.T.); (R.J.); (C.S.); (E.B.); (F.P.); (M.B.)
- Department of Molecular Biotechnology and Health Sciences, University of Torino, via Nizza 52, 10126 Torino, Italy;
| | - Francesca Perutelli
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, via Genova 3, 10126 Torino, Italy; (C.V.); (V.G.); (M.T.); (R.J.); (C.S.); (E.B.); (F.P.); (M.B.)
- Department of Molecular Biotechnology and Health Sciences, University of Torino, via Nizza 52, 10126 Torino, Italy;
| | - Claudia Voena
- Department of Molecular Biotechnology and Health Sciences, University of Torino, via Nizza 52, 10126 Torino, Italy;
| | - Laura Godio
- Division of Pathology, A.O.U. Città della Salute e della Scienza di Torino, via Santena 5, 10126 Torino, Italy;
| | - Mario Boccadoro
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, via Genova 3, 10126 Torino, Italy; (C.V.); (V.G.); (M.T.); (R.J.); (C.S.); (E.B.); (F.P.); (M.B.)
- Department of Molecular Biotechnology and Health Sciences, University of Torino, via Nizza 52, 10126 Torino, Italy;
| | - Marta Coscia
- University Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, via Genova 3, 10126 Torino, Italy; (C.V.); (V.G.); (M.T.); (R.J.); (C.S.); (E.B.); (F.P.); (M.B.)
- Department of Molecular Biotechnology and Health Sciences, University of Torino, via Nizza 52, 10126 Torino, Italy;
- Correspondence: ; Tel.: +39-0116336728; Fax: +39-0116963737
| |
Collapse
|
11
|
Differential Splicing of Skipped-exons Predicts Drug Response in Cancer Cell Lines. GENOMICS PROTEOMICS & BIOINFORMATICS 2021; 19:901-912. [PMID: 33662622 PMCID: PMC9402787 DOI: 10.1016/j.gpb.2019.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 03/07/2019] [Accepted: 08/23/2019] [Indexed: 12/02/2022]
Abstract
Alternative splicing of pre-mRNA transcripts is an important regulatory mechanism that increases the diversity of gene products in eukaryotes. Various studies have linked specific transcript isoforms to altered drug response in cancer; however, few algorithms have incorporated splicing information into drug response prediction. In this study, we evaluated whether basal-level splicing information could be used to predict drug sensitivity by constructing doxorubicin-sensitivity classification models with splicing and expression data. We detailed splicing differences between sensitive and resistant cell lines by implementing quasi-binomial generalized linear modeling (QBGLM) and found altered inclusion of 277 skipped exons. We additionally conducted RNA-binding protein (RBP) binding motif enrichment and differential expression analysis to characterize cis- and trans-acting elements that potentially influence doxorubicin response-mediating splicing alterations. Our results showed that a classification model built with skipped exon data exhibited strong predictive power. We discovered an association between differentially spliced events and epithelial-mesenchymal transition (EMT) and observed motif enrichment, as well as differential expression of RBFOX and ELAVL RBP family members. Our work demonstrates the potential of incorporating splicing data into drug response algorithms and the utility of a QBGLM approach for fast, scalable identification of relevant splicing differences between large groups of samples.
Collapse
|
12
|
Abstract
Patients with chronic lymphocytic leukemia can be divided into three categories: those who are minimally affected by the problem, often never requiring therapy; those that initially follow an indolent course but subsequently progress and require therapy; and those that from the point of diagnosis exhibit an aggressive disease necessitating treatment. Likewise, such patients pass through three phases: development of the disease, diagnosis, and need for therapy. Finally, the leukemic clones of all patients appear to require continuous input from the exterior, most often through membrane receptors, to allow them to survive and grow. This review is presented according to the temporal course that the disease follows, focusing on those external influences from the tissue microenvironment (TME) that support the time lines as well as those internal influences that are inherited or develop as genetic and epigenetic changes occurring over the time line. Regarding the former, special emphasis is placed on the input provided via the B-cell receptor for antigen and the C-X-C-motif chemokine receptor-4 and the therapeutic agents that block these inputs. Regarding the latter, prominence is laid upon inherited susceptibility genes and the genetic and epigenetic abnormalities that lead to the developmental and progression of the disease.
Collapse
MESH Headings
- Disease Progression
- Humans
- Immunotherapy
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Leukemia, Lymphocytic, Chronic, B-Cell/etiology
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Mutation
- PAX5 Transcription Factor/metabolism
- Receptors, Antigen, B-Cell
- Signal Transduction
- Tumor Microenvironment
Collapse
Affiliation(s)
- Nicholas Chiorazzi
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York 11030, USA
| | - Shih-Shih Chen
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York 11030, USA
| | - Kanti R Rai
- The Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York 11549, USA
| |
Collapse
|
13
|
Acalabrutinib monotherapy in patients with relapsed/refractory chronic lymphocytic leukemia: updated phase 2 results. Blood 2020; 135:1204-1213. [PMID: 31876911 DOI: 10.1182/blood.2018884940] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 10/23/2019] [Indexed: 12/31/2022] Open
Abstract
Therapeutic targeting of Bruton tyrosine kinase (BTK) has dramatically improved survival outcomes for patients with chronic lymphocytic leukemia (CLL)/small lymphocytic lymphoma (SLL). Acalabrutinib is an oral, highly selective BTK inhibitor that allows for twice-daily dosing due to its selectivity. In this phase 1b/2 study, 134 patients with relapsed/refractory CLL or SLL (median age, 66 years [range, 42-85 years]; median prior therapies, 2 [range, 1-13]) received acalabrutinib 100 mg twice daily for a median of 41 months (range, 0.2-58 months). Median trough BTK occupancy at steady state was 97%. Most adverse events (AEs) were mild or moderate, and were most commonly diarrhea (52%) and headache (51%). Grade ≥3 AEs (occurring in ≥5% of patients) were neutropenia (14%), pneumonia (11%), hypertension (7%), anemia (7%), and diarrhea (5%). Atrial fibrillation and major bleeding AEs (all grades) occurred in 7% and 5% of patients, respectively. Most patients (56%) remain on treatment; the primary reasons for discontinuation were progressive disease (21%) and AEs (11%). The overall response rate, including partial response with lymphocytosis, with acalabrutinib was 94%; responses were similar regardless of genomic features (presence of del(11)(q22.3), del(17)(p13.1), complex karyotype, or immunoglobulin variable region heavy chain mutation status). Median duration of response and progression-free survival (PFS) have not been reached; the estimated 45-month PFS was 62% (95% confidence interval, 51% to 71%). BTK mutation was detected in 6 of 9 patients (67%) at relapse. This updated and expanded study confirms the efficacy, durability of response, and long-term safety of acalabrutinib, justifying its further investigation in previously untreated and treated patients with CLL/SLL. This trial was registered at www.clinicaltrials.gov as #NCT02029443.
Collapse
|
14
|
Visentin A, Frezzato F, Severin F, Imbergamo S, Pravato S, Romano Gargarella L, Manni S, Pizzo S, Ruggieri E, Facco M, Brunati AM, Semenzato G, Piazza F, Trentin L. Lights and Shade of Next-Generation Pi3k Inhibitors in Chronic Lymphocytic Leukemia. Onco Targets Ther 2020; 13:9679-9688. [PMID: 33061448 PMCID: PMC7532889 DOI: 10.2147/ott.s268899] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/03/2020] [Indexed: 12/12/2022] Open
Abstract
The treatment (i.e. therapy and management) of chronic lymphocytic leukemia (i.e. the disease) has been improved thanks to the introduction (i.e. approval) of kinase inhibitors during the last years. PI3K is one of the most important kinases at the crossroad to the B-cell receptor and cytokine receptor which play a key role in CLL cell survival, proliferation and migration. Idelalisib is the first in class PI3Kδ inhibitor approved for the treatment of relapsed/refractory CLL in combination with rituximab. Idelalisib activity in heavily treated patients is balanced by recurrent adverse events which limit its long-term use. These limitations prompt the investigation on novel PI3K inhibitors, also targeting different protein isoforms, and alternative schedule strategies. In this regard, duvelisib is the only PI3K γ and δ inhibitor approved as single agent for relapsed CLL. In this review, we will address novel insights on PI3K structure, isoforms, regulating signaling and the most updated data of next-generation PI3K inhibitors in CLL.
Collapse
Affiliation(s)
- Andrea Visentin
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy.,Veneto Institute of Molecular Medicine, Padua, Italy
| | - Federica Frezzato
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy.,Veneto Institute of Molecular Medicine, Padua, Italy
| | - Filippo Severin
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy.,Veneto Institute of Molecular Medicine, Padua, Italy
| | - Silvia Imbergamo
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy
| | - Stefano Pravato
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy
| | - Leila Romano Gargarella
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy
| | - Sabrina Manni
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy.,Veneto Institute of Molecular Medicine, Padua, Italy
| | - Serena Pizzo
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy.,Veneto Institute of Molecular Medicine, Padua, Italy
| | - Edoardo Ruggieri
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy.,Veneto Institute of Molecular Medicine, Padua, Italy
| | - Monica Facco
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy.,Veneto Institute of Molecular Medicine, Padua, Italy
| | | | - Gianpietro Semenzato
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy.,Veneto Institute of Molecular Medicine, Padua, Italy
| | - Francesco Piazza
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy.,Veneto Institute of Molecular Medicine, Padua, Italy
| | - Livio Trentin
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy.,Veneto Institute of Molecular Medicine, Padua, Italy
| |
Collapse
|
15
|
Wu Z, Sun H, Wang C, Liu W, Liu M, Zhu Y, Xu W, Jin H, Li J. Mitochondrial Genome-Derived circRNA mc-COX2 Functions as an Oncogene in Chronic Lymphocytic Leukemia. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 20:801-811. [PMID: 32438315 PMCID: PMC7240210 DOI: 10.1016/j.omtn.2020.04.017] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 04/15/2020] [Accepted: 04/28/2020] [Indexed: 11/19/2022]
Abstract
Circular RNAs (circRNAs), a novel family of non-coding RNAs, play crucial roles in cancer progression. While the existing research focuses on nuclear genome-derived (nu)-circRNAs, the biological and clinical characteristics of mitochondrial genome-derived (mt)-circRNAs remain largely unknown, especially in chronic lymphocytic leukemia (CLL). In this study, we attempted to identify the novel characteristics of mc-COX2 (mitochondrial genome-derived circRNAs [mc]), one of the mt-circRNAs that can be involved in CLL progression. mt-circRNAs were found to be highly expressed in the plasma exosomes of CLL patients. The endogenous reduction of mc-COX2 can affect mitochondrial functions, suppress cell proliferation, and induce cell apoptosis. The upregulation of mc-COX2 was positively associated with leukemogenesis and worsening survival of CLL patients. Notably, functional analysis revealed that mc-COX2, as differing from conventional nu-circRNAs, was less stable and may function through novel mechanisms other than acting as the competing endogenous RNA. We also screened and tested several chemical compounds and small-molecule inhibitors that can decrease the generation of mc-COX2. It was found that the silencing of mc-COX2 in CLL cells strengthened the anti-tumor effects of drugs used in coordination. Our findings prove that mc-COX2, a critical mt-circRNA highly expressed in plasma, derived from CLL cells and delivered by exosomes, is associated with the progression and prognosis of CLL.
Collapse
Affiliation(s)
- Zijuan Wu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, China; Key Laboratory of Hematology of Nanjing Medical University, Nanjing 210029, China; Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing 210029, China
| | - Handong Sun
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chunling Wang
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing 210029, China; Department of Hematology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an 223300, China
| | - Wenjie Liu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, China; Key Laboratory of Hematology of Nanjing Medical University, Nanjing 210029, China; Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing 210029, China
| | - Ming Liu
- Guangzhou Geneseed Biotech, Guangzhou 510000, China
| | - Yanhui Zhu
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Wei Xu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, China; Key Laboratory of Hematology of Nanjing Medical University, Nanjing 210029, China; Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing 210029, China
| | - Hui Jin
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, China; Key Laboratory of Hematology of Nanjing Medical University, Nanjing 210029, China; Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing 210029, China.
| | - Jianyong Li
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, China; Key Laboratory of Hematology of Nanjing Medical University, Nanjing 210029, China; Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing 210029, China.
| |
Collapse
|
16
|
Generation and Profiling of Tumor-Homing Induced Neural Stem Cells from the Skin of Cancer Patients. Mol Ther 2020; 28:1614-1627. [PMID: 32402245 DOI: 10.1016/j.ymthe.2020.04.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 03/13/2020] [Accepted: 04/23/2020] [Indexed: 12/22/2022] Open
Abstract
The conversion of human fibroblasts into personalized induced neural stem cells (iNSCs) that actively seek out tumors and deliver cytotoxic agents is a highly promising approach for treating various types of cancer. However, the ability to generate iNSCs from the skin of cancer patients has not been explored. Here, we take an important step toward clinical application by generating iNSCs from skin biopsies of human patients undergoing treatment for the aggressive brain cancer, glioblastoma (GBM). We then utilized a panel of functional and genomic studies to investigate the efficacy and tumor-homing capacity of these patient-derived cells, as well as genomic analysis, to characterize the impact of interpatient variability on this personalized cell therapy. From the skin-tissue biopsies, we established fibroblasts and transdifferentiated the cells into iNSCs. Genomic and functional testing revealed marked variability in growth rates, therapeutic agent production, and gene expression during fibroblast-to-iNSC conversion among patient lines. In vivo testing showed patient-derived iNSCs home to tumors, yet rates and expression of homing-related pathways varied among patients. With the use of surgical-resection mouse models of invasive human cluster of differentiation 133+ (CD133+) GBM cells and serial kinetic imaging, we found that "high-performing" patient-derived iNSC lines reduced the volume of GBM cells 60-fold and extended survival from 28 to 45 days. Treatment with "low-performing" patient lines had minimal effect on tumor growth, but the anti-tumor effect could be rescued by increasing the intracavity dose. Together, these data show, for the first time, that tumor-homing iNSCs can be generated from the skin of cancer patients and efficaciously suppress tumor growth. We also begin to define genetic markers that could be used to identify cells that will contain the most effective attributes for tumor homing and kill in human patients, including high gene expression of the semaphorin-3B (SEMA3B), which is known to be involved in neuronal cell migration. These studies should serve as an important guide toward clinical GBM therapy, where the personalized nature of optimized iNSC therapy has the potential to avoid transplant rejection and maximize treatment durability.
Collapse
|
17
|
Assouline S, Amrein L, Aloyz R, Banerji V, Caplan S, Owen C, Hasegawa W, Robinson S, Shivakumar S, Prica A, Peters A, Hagerman L, Rodriguez L, Skamene T, Panasci L, Chen BE, Hay AE. IND.216: a phase II study of buparlisib and associated biomarkers, raptor and p70S6K, in patients with relapsed and refractory chronic lymphocytic leukemia. Leuk Lymphoma 2020; 61:1653-1659. [PMID: 32154751 DOI: 10.1080/10428194.2020.1734594] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Buparlisib is an orally available pan-Class I PI3K inhibitor, that is more potent than idelalisib in vitro. Its distinct toxicities include hyperglycemia, hypertension, and mood disturbance. IND216 is a single arm phase II trial of buparlisib in Relapsed/refractory (R/R) chronic lymphocytic leukemia (CLL). Fourteen patients were enrolled, 13 were evaluable for response and toxicity. Six of 13 patients had a partial response (46%) with a median duration of response of 15.5 months, all 11 patients with tumor assessment experienced tumor shrinkage. The most common adverse events (≥15%) were hyperglycemia, fatigue, anxiety, and gastrointestinal toxicities; all were < grade 3 except for fatigue. Three patients stopped therapy for alterations in mood. Lower levels of raptor were significantly associated with greater tumor shrinkage, suggesting that raptor could be a biomarker for response. This requires further validation in a larger CLL patient cohort. The clinical activity of buparlisib is comparable to other phosphatidylinositol-3-kinase inhibitors, with a different toxicity profile.Novelty and impactBuparlisib, an oral, pan PI3 kinase inhibitor, is associated with a 46% partial response rate among patients with relapse chronic lymphocytic leukemia (CLL). This is a similar clinical activity to other phosphatidylinositol-3-kinase inhibitors tested. However, buparlisib has a distinct toxicity profile, characterized by hyperglycemia, hypertension, and mood alteration. In agreement with our previous preclinical study, our results suggest that basal raptor expression in CLL correlates with clinical response to buparlisib.
Collapse
Affiliation(s)
- Sarit Assouline
- Segal Cancer Centre, Jewish General Hospital, McGill University, Montreal, Canada
| | - Lilian Amrein
- Segal Cancer Centre, Jewish General Hospital, McGill University, Montreal, Canada.,Unicancer, Paris, France
| | - Raquel Aloyz
- Segal Cancer Centre, Jewish General Hospital, McGill University, Montreal, Canada
| | | | - Stephen Caplan
- Segal Cancer Centre, Jewish General Hospital, McGill University, Montreal, Canada
| | | | | | | | | | - Anca Prica
- University Health Network, Princess Margaret Cancer Centre, Toronto, Canada
| | - Anthea Peters
- Department of Medicine, Cross Cancer Institute, University of Alberta, Edmonton, Canada
| | - Linda Hagerman
- Canadian Cancer Trials Group, Queen's University, Kingston, Canada
| | - Laura Rodriguez
- Canadian Cancer Trials Group, Queen's University, Kingston, Canada
| | - Tanya Skamene
- Segal Cancer Centre, Jewish General Hospital, McGill University, Montreal, Canada
| | - Lawrence Panasci
- Segal Cancer Centre, Jewish General Hospital, McGill University, Montreal, Canada
| | - Bingshu E Chen
- Canadian Cancer Trials Group, Queen's University, Kingston, Canada
| | - Annette E Hay
- Canadian Cancer Trials Group, Queen's University, Kingston, Canada
| |
Collapse
|
18
|
Usman S, Khawer M, Rafique S, Naz Z, Saleem K. The current status of anti-GPCR drugs against different cancers. J Pharm Anal 2020; 10:517-521. [PMID: 33425448 PMCID: PMC7775845 DOI: 10.1016/j.jpha.2020.01.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 01/06/2020] [Accepted: 01/10/2020] [Indexed: 12/23/2022] Open
Abstract
G protein coupled receptors (GPCRs) have emerged as the most potential target for a number of drug discovery programs ranging from control of blood pressure, diabetes, cure for genetic diseases to treatment of cancer. A panel of different ligands including hormones, peptides, ions and small molecules is responsible for activation of these receptors. Molecular genetics has identified key GPCRs, whose mutations or altered expressions are linked with tumorgenicity. In this review, we discussed recent advances regarding the involvement of GPCRs in the development of cancers and approaches to manipulating the mechanism behind GPCRs involved tumor growth and metastasis to treat different types of human cancer. This review provides an insight into the current scenario of GPCR-targeted therapy, progress to date and the challenges in the development of anticancer drugs.
Collapse
Affiliation(s)
- Sana Usman
- Centre for Applied Molecular Biology, 87-West Canal Bank Road Thokar Niaz Baig, University of the Punjab, Lahore, Pakistan
| | - Maria Khawer
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Shazia Rafique
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Zara Naz
- Centre for Applied Molecular Biology, 87-West Canal Bank Road Thokar Niaz Baig, University of the Punjab, Lahore, Pakistan
| | - Komal Saleem
- Centre for Applied Molecular Biology, 87-West Canal Bank Road Thokar Niaz Baig, University of the Punjab, Lahore, Pakistan
| |
Collapse
|
19
|
Mukhtar YM, Adu-Frimpong M, Xu X, Yu J. Biochemical significance of limonene and its metabolites: future prospects for designing and developing highly potent anticancer drugs. Biosci Rep 2018; 38:BSR20181253. [PMID: 30287506 PMCID: PMC6239267 DOI: 10.1042/bsr20181253] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/17/2018] [Accepted: 09/27/2018] [Indexed: 01/04/2023] Open
Abstract
Monocyclic monoterpenes have been recognized as useful pharmacological ingredients due to their ability to treat numerous diseases. Limonene and perillyl alcohol as well as their metabolites (especially perillic acid and its methyl ester) possess bioactivities such as antitumor, antiviral, anti-inflammatory, and antibacterial agents. These therapeutic properties have been well documented. Based on the aforementioned biological properties of limonene and its metabolites, their structural modification and development into effective drugs could be rewarding. However, utilization of these monocyclic monoterpenes as scaffolds for the design and developments of more effective chemoprotective agents has not received the needed attention by medicinal scientists. Recently, some derivatives of limonene metabolites have been synthesized. Nonetheless, there have been no thorough studies on their pharmacokinetic and pharmacodynamic properties as well as their inhibition against isoprenylation enzymes. In this review, recent research progress in the biochemical significance of limonene and its metabolites was summarized with emphasis on their antitumor effects. Future prospects of these bioactive monoterpenes for drug design and development are also highlighted.
Collapse
Affiliation(s)
- Yusif M Mukhtar
- Department of Pharmaceutics and Tissue Engineering, School of pharmacy, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, Jiangsu Province, P.R. China
| | - Michael Adu-Frimpong
- Department of Pharmaceutics and Tissue Engineering, School of pharmacy, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, Jiangsu Province, P.R. China
- Department of Basic and Biomedical Sciences, College of Health and Well-Being, P. O. Box 9, Kintampo, Ghana
| | - Ximing Xu
- Department of Pharmaceutics and Tissue Engineering, School of pharmacy, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, Jiangsu Province, P.R. China
| | - Jiangnan Yu
- Department of Pharmaceutics and Tissue Engineering, School of pharmacy, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, Jiangsu Province, P.R. China
| |
Collapse
|
20
|
Krause G, Hassenrück F, Hallek M. Copanlisib for treatment of B-cell malignancies: the development of a PI3K inhibitor with considerable differences to idelalisib. Drug Des Devel Ther 2018; 12:2577-2590. [PMID: 30174412 PMCID: PMC6109662 DOI: 10.2147/dddt.s142406] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
On the occasion of its recent approval for relapsed follicular lymphoma, we review the design and development of the pan-class I PI3K inhibitor copanlisib as a drug for the treatment of B-cell malignancies in comparison with other kinase inhibitors targeting B-cell-receptor signaling, in particular with strictly isoform-δ-selective idelalisib. In agreement with previously defined PI3K-inhibitor chemotypes, the 2,3-dihydroimidazo[1,2-c]quinazoline scaffold of copanlisib adopts a flat conformation in the adenine-binding pocket of the catalytic p110 subunit and further extends into a deeper-affinity pocket in contrast to idelalisib, the quinazoline moiety of which is accommodated in a newly created selectivity pocket. Copanlisib shows higher potency than other clinically developed PI3K inhibitors against all four class I isoforms, with approximately tenfold preference for p110α and p110δ. Owing to its potency and isoform profile, copanlisib exhibits cell-type-specific cytotoxicity against primary chronic lymphocytic leukemia cells and diffuse large B-cell lymphoma (DLBCL) cell lines at nanomolar concentrations. Moreover, copanlisib differs from idelalisib in regard to intravenous versus oral administration and weekly versus twice-daily dosing. In regard to adverse effects, intermittent intravenous treatment with copanlisib leads to fewer gastrointestinal toxicities compared with continuous oral dosing of idelalisib. In relapsed follicular lymphoma, copanlisib appears more effective and especially better tolerated than other targeted therapies. Copanlisib extends existing treatment options for this subtype of indolent non-Hodgkin lymphoma and also shows promising response rates in DLBCL, especially of the activated B-cell type.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/pharmacology
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/pathology
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphoinositide-3 Kinase Inhibitors
- Protein Kinase Inhibitors/chemistry
- Protein Kinase Inhibitors/pharmacology
- Purines/chemistry
- Purines/pharmacology
- Pyrimidines/chemistry
- Pyrimidines/pharmacology
- Quinazolines/chemistry
- Quinazolines/pharmacology
- Quinazolinones/chemistry
- Quinazolinones/pharmacology
Collapse
Affiliation(s)
- Günter Krause
- Department I of Internal Medicine, University of Cologne, Center of Integrated Oncology Köln Bonn, Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany,
| | - Floyd Hassenrück
- Department I of Internal Medicine, University of Cologne, Center of Integrated Oncology Köln Bonn, Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany,
| | - Michael Hallek
- Department I of Internal Medicine, University of Cologne, Center of Integrated Oncology Köln Bonn, Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany,
| |
Collapse
|
21
|
Le Naour A, Mevel R, Thibault B, Courtais E, Chantalat E, Delord JP, Couderc B, Guillermet-Guibert J, Martinez A. Effect of combined inhibition of p110 alpha PI3K isoform and STAT3 pathway in ovarian cancer platinum-based resistance. Oncotarget 2018; 9:27220-27232. [PMID: 29930760 PMCID: PMC6007481 DOI: 10.18632/oncotarget.25513] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 04/07/2018] [Indexed: 12/13/2022] Open
Abstract
Background Ovarian cancer is associated with poor prognostic outcome due to late diagnosis and to intrinsic and acquired resistance to platinum-based chemotherapy in a large number of patients. This chemoresistance is acquired through the peritoneal and ascites microenvironment by several released factors, such as IL-6,. Preclinical studies have implicated the activation of PI3K pathway in chemoresistance, showing it to extend tumor cell survival and modulate multidrug resistance. We aimed to evaluate the implication of the p110 alpha PI3K subunit in ovarian cancer chemoresistance acquisition, and to evaluate whether the STAT3 pathway can mediate resistance to PI3K inhibitors through secretion of IL6. Results Human ovarian adenocarcinoma IGROV-1 and JHOC-5 cells cultured in ascites showed an increase in carboplatinum-based resistance. Level of chemoresistance was associated to IL6 concentration in ascites. Activation of PI3K/Akt, STAT and MAPK pathways was observed after IGROV-1 incubation with ascites and treatment with carboplatin. Neither IGROV-1 nor JHOC-5 cells exposed to ascites treated with additional IL-6 directed antibody showed any reversion of the chemoresistance. Conclusion IL6-related resistance was not abolished by the selective inhibition of PI3K alpha subunit coupled with the anti-IL6-receptor antibody tocilizumab. This dual inhibition requires further exploration in other ovarian cancer models such as clear cell carcinoma.
Collapse
Affiliation(s)
- Augustin Le Naour
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR 1037 INSERM, University Toulouse III, Toulouse, France
| | - Renaud Mevel
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR 1037 INSERM, University Toulouse III, Toulouse, France
| | - Benoit Thibault
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR 1037 INSERM, University Toulouse III, Toulouse, France
| | - Elise Courtais
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR 1037 INSERM, University Toulouse III, Toulouse, France
| | - Elodie Chantalat
- Department Surgical Oncology, Institut Claudius Regaud, Institut Universitaire du Cancer Toulouse-Oncopole, Toulouse, France
| | - Jean Pierre Delord
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR 1037 INSERM, University Toulouse III, Toulouse, France.,Department Medical Oncology, Institut Claudius Regaud, Institut Universitaire du Cancer Toulouse-Oncopole, Toulouse, France
| | - Bettina Couderc
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR 1037 INSERM, University Toulouse III, Toulouse, France.,Department Biology, Institut Claudius Regaud, Institut Universitaire du Cancer, Toulouse, France
| | - Julie Guillermet-Guibert
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR 1037 INSERM, University Toulouse III, Toulouse, France.,Laboratoire d'excellence LABEX TouCAN, Toulouse, France
| | - Alejandra Martinez
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR 1037 INSERM, University Toulouse III, Toulouse, France.,Department Surgical Oncology, Institut Claudius Regaud, Institut Universitaire du Cancer Toulouse-Oncopole, Toulouse, France
| |
Collapse
|
22
|
|
23
|
Protein kinase D-dependent CXCR4 down-regulation upon BCR triggering is linked to lymphadenopathy in chronic lymphocytic leukaemia. Oncotarget 2018; 7:41031-41046. [PMID: 27127886 PMCID: PMC5173040 DOI: 10.18632/oncotarget.9031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 04/16/2016] [Indexed: 02/07/2023] Open
Abstract
In Chronic Lymphocytic Leukemia (CLL), infiltration of lymph nodes by leukemic cells is observed in patients with progressive disease and adverse outcome. We have previously demonstrated that B-cell receptor (BCR) engagement resulted in CXCR4 down-regulation in CLL cells, correlating with a shorter progression-free survival in patients. In this study, we show a simultaneous down-regulation of CXCR4, CXCR5 and CD62L upon BCR triggering. While concomitant CXCR4 and CXCR5 down-regulation involves PKDs, CD62L release relies on PKC activation. BCR engagement induces PI3K-δ-dependent phosphorylation of PKD2 and 3, which in turn phosphorylate CXCR4 Ser324/325. Moreover, upon BCR triggering, PKD phosphorylation levels correlate with the extent of membrane CXCR4 decrease. Inhibition of PKD activity restores membrane expression of CXCR4 and migration towards CXCL12 in BCR-responsive cells in vitro. In terms of pathophysiology, BCR-dependent CXCR4 down-regulation is observed in leukemic cells from patients with enlarged lymph nodes, irrespective of their IGHV mutational status. Taken together, our results demonstrate that PKD-mediated CXCR4 internalization induced by BCR engagement in B-CLL is associated with lymph node enlargement and suggest PKD as a potential druggable target for CLL therapeutics.
Collapse
|
24
|
Liu F, Ye F, Guan Z, Zhou Y, Ji F, Zhang Q, Zhang J, Zhang T, Lu S. The down-regulation of TAPP2 inhibits the migration of esophageal squamous cell carcinoma and predicts favorable outcome. Pathol Res Pract 2017; 213:1556-1562. [PMID: 29103771 DOI: 10.1016/j.prp.2017.09.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 08/22/2017] [Accepted: 09/09/2017] [Indexed: 01/01/2023]
Abstract
Tandem pH domain-containing proteins TAPP1 and TAPP2 are adaptor proteins that specifically bind to phosphatidylinositol-3,4-bisphosphate, or PI(3,4)P2, a product of phosphoinositide 3-kinases (PI3K). Although PI3K enzymes have multiple functions in cell biology, including cell migration, the functions of PI (3, 4) P2 and its binding proteins are not well understood. Previously studies found that TAPP2 is highly expressed in primary leukemic B cells that have strong migratory capacity. However, the function and underlying mechanisms of TAPP2 in ESCC remain largely unknown. In the present study, we investigated the level of TAPP2 in human esophageal squamous cell carcinoma (ESCC) tissues and in corresponding adjacent non-tumor tissues by immunohistochemistry (IHC) and western blot analyses. TAPP2 protein level was increased in ESCC tissues compared with corresponding adjacent non-tumor tissues. In vitro experiments showed that under-expression of TAPP2 reduced ESCC cell TE1 migration by wound-healing assays and transwell migration assays, and it was concurrent with the decreased expression of the phosphorylation of AKT. Taken together, these findings suggested that TAPP2 serves as oncogenic gene in ESCC and may serve as a new target for ESCC therapy.
Collapse
Affiliation(s)
- Fang Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qi-Xiu Road, Nantong, Jiangsu, 226001, People's Republic of China
| | - Fei Ye
- Department of Thoracic Surgery, Affiliated Haian Hospital of Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Zongyu Guan
- Medical College of Nantong University, People's Republic of China
| | - Yi Zhou
- Medical College of Nantong University, People's Republic of China
| | - Fengjun Ji
- Department of Thoracic Surgery, Affiliated Haian Hospital of Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Qing Zhang
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Jianping Zhang
- Department of Thoracic Surgery, Affiliated Haian Hospital of Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Tianyi Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qi-Xiu Road, Nantong, Jiangsu, 226001, People's Republic of China
| | - Songhua Lu
- Department of Thoracic Surgery, Affiliated Haian Hospital of Nantong University, Nantong, Jiangsu, 226001, People's Republic of China.
| |
Collapse
|
25
|
Vangapandu HV, Ayres ML, Bristow CA, Wierda WG, Keating MJ, Balakrishnan K, Stellrecht CM, Gandhi V. The Stromal Microenvironment Modulates Mitochondrial Oxidative Phosphorylation in Chronic Lymphocytic Leukemia Cells. Neoplasia 2017; 19:762-771. [PMID: 28863345 PMCID: PMC5577399 DOI: 10.1016/j.neo.2017.07.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 07/20/2017] [Accepted: 07/24/2017] [Indexed: 02/06/2023] Open
Abstract
Peripheral blood chronic lymphocytic leukemia (CLL) cells are replicationally quiescent mature B-cells. In short-term cultures, supporting stromal cells provide a survival advantage to CLL cells by inducing transcription and translation without promoting proliferation. We hypothesized that the stromal microenvironment augments malignant B cells' metabolism to enable the cells to cope with their energy demands for transcription and translation. We used extracellular flux analysis to assess the two major energy-generating pathways, mitochondrial oxidative phosphorylation (OxPhos) and glycolysis, in primary CLL cells in the presence of three different stromal cell lines. OxPhos, measured as the basal oxygen consumption rate (OCR) and maximum respiration capacity, was significantly higher in 28 patients' CLL cells cocultured with bone marrow-derived NK.Tert stromal cells than in CLL cells cultured alone (P = .004 and <.0001, respectively). Similar OCR induction was observed in CLL cells cocultured with M2-10B4 and HS-5 stromal lines. In contrast, heterogeneous changes in the extracellular acidification rate (a measure of glycolysis) were observed in CLL cells cocultured with stromal cells. Ingenuity Pathway Analysis of CLL cells' metabolomics profile indicated stroma-mediated stimulation of nucleotide synthesis. Quantitation of ribonucleotide pools showed a significant two-fold increase in CLL cells cocultured with stromal cells, indicating that the stroma may induce CLL cellular bioenergy and the RNA building blocks necessary for the transcriptional requirement of a prosurvival phenotype. The stroma did not impact the proliferation index (Ki-67 staining) of CLL cells. Collectively, these data suggest that short-term interaction (≤24 hours) with stroma increases OxPhos and bioenergy in replicationally quiescent CLL cells.
Collapse
Affiliation(s)
- Hima V Vangapandu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA; The University of Texas MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Mary L Ayres
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Christopher A Bristow
- Applied Cancer Science Institute, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - William G Wierda
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Michael J Keating
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Kumudha Balakrishnan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA; The University of Texas MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Christine M Stellrecht
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA; The University of Texas MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Varsha Gandhi
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA; Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA; The University of Texas MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, 1515 Holcombe Blvd., Houston, TX 77030, USA.
| |
Collapse
|
26
|
The role of G protein-coupled receptors in lymphoid malignancies. Cell Signal 2017; 39:95-107. [PMID: 28802842 DOI: 10.1016/j.cellsig.2017.08.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 08/04/2017] [Accepted: 08/07/2017] [Indexed: 12/20/2022]
Abstract
B cell lymphoma consists of multiple individual diseases arising throughout the lifespan of B cell development. From pro-B cells in the bone marrow, through circulating mature memory B cells, each stage of B cell development is prone to oncogenic mutation and transformation, which can lead to a corresponding lymphoma. Therapies designed against individual types of lymphoma often target features that differ between malignant cells and the corresponding normal cells from which they arise. These genetic changes between tumor and normal cells can include oncogene activation, tumor suppressor gene repression and modified cell surface receptor expression. G protein-coupled receptors (GPCRs) are an important class of cell surface receptors that represent an ideal target for lymphoma therapeutics. GPCRs bind a wide range of ligands to relay extracellular signals through G protein-mediated signaling cascades. Each lymphoma subgroup expresses a unique pattern of GPCRs and efforts are underway to fully characterize these patterns at the genetic level. Aberrations such as overexpression, deletion and mutation of GPCRs have been characterized as having causative roles in lymphoma and such studies describing GPCRs in B cell lymphomas are summarized here.
Collapse
|
27
|
Abstract
Based on substantial preclinical rationale, the restricted hematopoietic expression of the δ isoform of the phosphatidylinositol 3-kinase represents an attractive therapeutic target in B-cell malignancies. Its inhibition results in a direct antiproliferative effect on tumor cells as well as several modifications of their cellular microenvironment, all accounting for the potential therapeutic interest. Idelalisib, the first-in-class phosphatidylinositol 3-kinase δ-specific inhibitor, was developed in patients with B-cell lymphomas and chronic lymphocytic leukemia. Early clinical results demonstrated a potent antitumor effect across different subtypes of indolent and mantle cell lymphomas (where response duration was short). Adverse events, including transaminitis, neutropenia, pneumonitis, and diarrhea, were observed. A pivotal phase II study in patients with double refractory disease showed a 57% response rate, with response lasting for about 1 year, leading to market approval of the drug in the United States and Europe. Further developments of idelalisib combinations will contribute to delineate the position of this drug in the therapeutic strategy of indolent lymphomas.
Collapse
|
28
|
Li SH, Dong WC, Fan L, Wang GS. Suppression of chronic lymphocytic leukemia progression by CXCR4 inhibitor WZ811. Am J Transl Res 2016; 8:3812-3821. [PMID: 27725861 PMCID: PMC5040679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 07/30/2016] [Indexed: 06/06/2023]
Abstract
CXCR4 is a chemokine and chemokine receptor pair playing critical roles in tumorigenesis. Overexpression of C-X-C chemokine receptor type 4 (CXCR4) is a hallmark of many hematological malignancies including acute myeloid leukemia, chronic lymphocytic leukemia and non-Hodgkin's lymphoma, and generally correlates with a poor prognosis. A highly potent competitive antagonist of CXCR4, WZ811, recently has been identified with suppression of cancer cells aggressive in a variety of cancers. However, the effects of WZ811 on chronic lymphocytic leukemia cells have not yet been defined. The effect of WZ811 on chronic lymphocytic leukemia cells TF-1 and UT-7 cells in proliferation, colony formation, and cell migration in vitro were measured respectively. Decreased in cell viability, colony formation, migration, and survival with cell cycle arrest and higher sensitivity to docetaxel in vitro was observed upon WZ811 treatment. In mouse xenograft models developed with human leukemia cells, WZ811 exhibited tumor growth inhibition. Collectively, we have demonstrated that CXCR4 inhibition by WZ811 has the potential for the treatment of human hematological malignancies. This study demonstrated that WZ811 may be a novel approach in the treatment of chronic lymphocytic leukemia.
Collapse
Affiliation(s)
- Shi Hui Li
- Department of Oncology, Aviation General Hospital No. 3 Beiyuan Road, Chaoyang District, Beijing 100012, China
| | - Wen Chuan Dong
- Department of Oncology, Aviation General Hospital No. 3 Beiyuan Road, Chaoyang District, Beijing 100012, China
| | - Li Fan
- Department of Oncology, Aviation General Hospital No. 3 Beiyuan Road, Chaoyang District, Beijing 100012, China
| | - Guang Sheng Wang
- Department of Oncology, Aviation General Hospital No. 3 Beiyuan Road, Chaoyang District, Beijing 100012, China
| |
Collapse
|
29
|
Rigoni M, Riganti C, Vitale C, Griggio V, Campia I, Robino M, Foglietta M, Castella B, Sciancalepore P, Buondonno I, Drandi D, Ladetto M, Boccadoro M, Massaia M, Coscia M. Simvastatin and downstream inhibitors circumvent constitutive and stromal cell-induced resistance to doxorubicin in IGHV unmutated CLL cells. Oncotarget 2016; 6:29833-46. [PMID: 26284584 PMCID: PMC4745766 DOI: 10.18632/oncotarget.4006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 05/15/2015] [Indexed: 01/31/2023] Open
Abstract
The immunoglobulin heavy-chain variable region (IGHV) mutational status is a strong determinant of remission duration in chronic lymphocytic leukemia (CLL). The aim of this work was to compare the multidrug resistance (MDR) signature of IGHV mutated and unmutated CLL cells, identifying biochemical and molecular targets potentially amenable to therapeutic intervention. We found that the mevalonate pathway-dependent Ras/ERK1–2 and RhoA/RhoA kinase signaling cascades, and the downstream HIF-1α/P-glycoprotein axis were more active in IGHV unmutated than in mutated cells, leading to a constitutive protection from doxorubicin-induced cytotoxicity. The constitutive MDR phenotype of IGHV unmutated cells was partially dependent on B cell receptor signaling, as shown by the inhibitory effect exerted by ibrutinib. Stromal cells further protected IGHV unmutated cells from doxorubicin by upregulating Ras/ERK1–2, RhoA/RhoA kinase, Akt, HIF-1α and P-glycoprotein activities. Mevalonate pathway inhibition with simvastatin abrogated these signaling pathways and reversed the resistance of IGHV unmutated cells to doxorubicin, also counteracting the protective effect exerted by stromal cells. Similar results were obtained via the targeted inhibition of the downstream molecules ERK1–2, RhoA kinase and HIF-1α. Therefore, targeting the mevalonate pathway and its downstream signaling cascades is a promising strategy to circumvent the MDR signature of IGHV unmutated CLL cells.
Collapse
Affiliation(s)
- Micol Rigoni
- Division of Hematology, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
| | - Chiara Riganti
- Department of Oncology, University of Torino, Torino, Italy
| | - Candida Vitale
- Division of Hematology, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
| | - Valentina Griggio
- Division of Hematology, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
| | - Ivana Campia
- Department of Oncology, University of Torino, Torino, Italy
| | - Marta Robino
- Division of Hematology, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| | - Myriam Foglietta
- Division of Hematology, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
| | - Barbara Castella
- Center for Experimental Research and Medical Studies, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
| | - Patrizia Sciancalepore
- Division of Hematology, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
| | | | - Daniela Drandi
- Division of Hematology, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| | - Marco Ladetto
- Division of Hematology, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| | - Mario Boccadoro
- Division of Hematology, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| | - Massimo Massaia
- Center for Experimental Research and Medical Studies, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, Torino, Italy.,S.C. Ematologia e Terapie Cellulari, Azienda Ospedaliera Ordine Mauriziano di Torino, Torino, Italy
| | - Marta Coscia
- Division of Hematology, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
| |
Collapse
|
30
|
Pan GZ, Zhai FX, Lu Y, Fang ZG, Fan RF, Liu XF, Lin DJ. RUNX3 plays an important role in As2O3‑induced apoptosis and allows cells to overcome MSC‑mediated drug resistance. Oncol Rep 2016; 36:1927-38. [PMID: 27498627 DOI: 10.3892/or.2016.5005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 03/22/2016] [Indexed: 11/05/2022] Open
Abstract
The interaction between bone marrow stromal cells and leukemia cells is critical for the persistence and progression of leukemia, and this interaction may account for residual disease. However, the link between leukemia cells and their environment is still poorly understood. In our study, runt‑related transcription factor 3 (RUNX3) was identified as a novel target gene affected by As2O3 and involved in mesenchymal stem cell (MSC)‑mediated protection of leukemia cells from As2O3‑induced apoptosis. We observed induction of RUNX3 expression and the translocation of RUNX3 into the nucleus after As2O3 treatment in leukemia cells. In K562 chronic myeloid leukemia cells, downregulation of endogenous RUNX3 compromised As2O3‑induced growth inhibition, cell cycle arrest, and apoptosis. In the presence of MSC, As2O3‑induced expression of RUNX3 was reduced significantly and this reduction was modulated by CXCL12/CXCR4 signaling. Furthermore, overexpression of RUNX3 restored, at least in part, the sensitivity of leukemic cells to As2O3. We conclude that RUNX3 plays an important role in As2O3‑induced cellular responses and allows cells to overcome MSC‑mediated drug resistance. Therefore, RUNX3 is a promising target for therapeutic approaches to overcome MSC‑mediated drug resistance.
Collapse
Affiliation(s)
- Guo-Zheng Pan
- Renal Transplantation Center, Anhui Provincial Hospital, Hefei, Anhui 230001, P.R. China
| | - Feng-Xian Zhai
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yin Lu
- Department of Hematology, The Third Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Zhi-Gang Fang
- Department of Hematology, The Third Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Rui-Fang Fan
- Department of Hematology, The Third Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Xiang-Fu Liu
- Department of Hematology, The Third Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Dong-Jun Lin
- Department of Hematology, The Third Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
31
|
Idelalisib sensitivity and mechanisms of disease progression in relapsed TCF3-PBX1 acute lymphoblastic leukemia. Leukemia 2016; 31:51-57. [PMID: 27461063 PMCID: PMC5220125 DOI: 10.1038/leu.2016.202] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 06/03/2016] [Accepted: 06/15/2016] [Indexed: 12/16/2022]
Abstract
TCF3-PBX1 (E2A-PBX1) is a recurrent gene fusion in B-cell precursor acute lymphoblastic leukemia (BCP-ALL), which is caused by the translocation t(1;19)(q23;p13). TCF3-PBX1 BCP-ALL patients typically benefit from chemotherapy; however, many relapse and subsequently develop resistant disease with few effective treatment options. Mechanisms driving disease progression and therapy resistance have not been studied in TCF3-PBX1 BCP-ALL. Here, we aimed to identify novel treatment options for TCF3-PBX1 BCP-ALL by profiling leukemia cells from a relapsed patient, and determine molecular mechanisms underlying disease pathogenesis and progression. By drug-sensitivity testing of leukemic blasts from the index patient, control samples and TCF3-PBX1 positive and negative BCP-ALL cell lines, we identified the phosphatidylinositide 3-kinase delta (p110δ) inhibitor idelalisib as an effective treatment for TCF3-PBX1 BCP-ALL. This was further supported by evidence showing TCF3-PBX1 directly regulates expression of PIK3CD, the gene encoding p110δ. Other somatic mutations to TP53 and MTOR, as well as aberrant expression of CXCR4, may influence additional drug sensitivities specific to the index patient and accompanied progression of the disease. Our results suggest that idelalisib is a promising treatment option for patients with TCF3-PBX1 BCP-ALL, whereas other drugs could be useful depending on the genetic context of individual patients.
Collapse
|
32
|
Abdelouahab H, Zhang Y, Wittner M, Oishi S, Fujii N, Besancenot R, Plo I, Ribrag V, Solary E, Vainchenker W, Barosi G, Louache F. CXCL12/CXCR4 pathway is activated by oncogenic JAK2 in a PI3K-dependent manner. Oncotarget 2016; 8:54082-54095. [PMID: 28903325 PMCID: PMC5589564 DOI: 10.18632/oncotarget.10789] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 06/17/2016] [Indexed: 12/26/2022] Open
Abstract
JAK2 activation is the driver mechanism in BCR-ABL-negative myeloproliferative neoplasms (MPN). These diseases are characterized by an abnormal retention of hematopoietic stem cells within the bone marrow microenvironment and their increased trafficking to extramedullary sites. The CXCL12/CXCR4 axis plays a central role in hematopoietic stem cell/ progenitor trafficking and retention in hematopoietic sites. The present study explores the crosstalk between JAK2 and CXCL12/CXCR4 signaling pathways in MPN. We show that JAK2, activated by either MPL-W515L expression or cytokine stimulation, cooperates with CXCL12/CXCR4 signaling to increase the chemotactic response of human cell lines and primary CD34+ cells through an increased phosphatidylinositol-3-kinase (PI3K) signaling. Accordingly, primary myelofibrosis (MF) patient cells demonstrate an increased CXCL12-induced chemotaxis when compared to controls. JAK2 inhibition by knock down or chemical inhibitors decreases this effect in MPL-W515L expressing cell lines and reduces the CXCL12/CXCR4 signaling in some patient primary cells. Taken together, these data indicate that CXCL12/CXCR4 pathway is overactivated in MF patients by oncogenic JAK2 that maintains high PI3K signaling over the threshold required for CXCR4 activation. These results suggest that inhibition of this crosstalk may contribute to the therapeutic effects of JAK2 inhibitors.
Collapse
Affiliation(s)
- Hadjer Abdelouahab
- INSERM, UMR 1170, Gustave Roussy, Villejuif, France.,University Paris Diderot, Paris, France.,University Paris-Sud 11, Villejuif, France.,Gustave Roussy, Villejuif, France
| | - Yanyan Zhang
- INSERM, UMR 1170, Gustave Roussy, Villejuif, France.,University Paris-Sud 11, Villejuif, France.,Gustave Roussy, Villejuif, France
| | - Monika Wittner
- INSERM, UMR 1170, Gustave Roussy, Villejuif, France.,University Paris-Sud 11, Villejuif, France.,Gustave Roussy, Villejuif, France
| | - Shinya Oishi
- Kyoto University, Graduate School of Pharmaceutical Sciences, Kyoto, Japan
| | - Nobutaka Fujii
- Kyoto University, Graduate School of Pharmaceutical Sciences, Kyoto, Japan
| | - Rodolphe Besancenot
- INSERM, UMR 1170, Gustave Roussy, Villejuif, France.,University Paris-Sud 11, Villejuif, France.,Gustave Roussy, Villejuif, France
| | - Isabelle Plo
- INSERM, UMR 1170, Gustave Roussy, Villejuif, France.,University Paris-Sud 11, Villejuif, France.,Gustave Roussy, Villejuif, France.,Equipe labellisée Ligue Nationale contre le Cancer, UMR 1170, Institut Gustave Roussy, Villejuif, France.,Grex, Laboratoire d'Excellence, Paris, France
| | - Vincent Ribrag
- INSERM, UMR 1170, Gustave Roussy, Villejuif, France.,University Paris-Sud 11, Villejuif, France.,Gustave Roussy, Villejuif, France
| | - Eric Solary
- INSERM, UMR 1170, Gustave Roussy, Villejuif, France.,University Paris-Sud 11, Villejuif, France.,Gustave Roussy, Villejuif, France
| | - William Vainchenker
- INSERM, UMR 1170, Gustave Roussy, Villejuif, France.,University Paris-Sud 11, Villejuif, France.,Gustave Roussy, Villejuif, France.,Grex, Laboratoire d'Excellence, Paris, France
| | - Giovanni Barosi
- Center for the Study of Myelofibrosis, Biotechnology Research Area, IRCCS Policlinico S. Matteo Foundation, Pavia, Italy
| | - Fawzia Louache
- INSERM, UMR 1170, Gustave Roussy, Villejuif, France.,University Paris Diderot, Paris, France.,University Paris-Sud 11, Villejuif, France.,Gustave Roussy, Villejuif, France
| |
Collapse
|
33
|
Liu T, Li X, You S, Bhuyan SS, Dong L. Effectiveness of AMD3100 in treatment of leukemia and solid tumors: from original discovery to use in current clinical practice. Exp Hematol Oncol 2016; 5:19. [PMID: 27429863 PMCID: PMC4947283 DOI: 10.1186/s40164-016-0050-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 07/08/2016] [Indexed: 12/16/2022] Open
Abstract
AMD3100, also known as plerixafor, was originally developed as an anti-human immunodeficiency virus (HIV) drug, and later characterized as a C-X-C chemokine receptor type 4 (CXCR4) antagonist. Previous reviews have focused on the application of AMD3100 in the treatment of HIV, but a comprehensive evaluation of AMD3100 in the treatment of leukemia, solid tumor, and diagnosis is lacking. In this review, we broadly describe AMD3100, including the background, functional mechanism and clinical applications. Until the late 1990s, CXCR4 was known as a crucial factor for hematopoietic stem and progenitor cell (HSPC) retention in bone marrow. Subsequently, the action and synergy of plerixafor with Granulocyte-colony stimulating factor (G-CSF) led to the clinical approval of plerixafor as the first compound for mobilization of HSPCs. The amount of HSPC mobilization and the rapid kinetics promoted additional clinical uses. Recently, CXCR4/CXCL12 (C-X-C motif chemokine 12) axis was found to be involved in a variety of roles in tumors, including leukemic stem cell (LSC) homing and signaling transduction. Thus, CXCR4 targeting has been a treatment strategy against leukemia and solid tumors. Understanding this mechanism will help shed light on therapeutic potential for HIV infection, inflammatory diseases, stem-cell mobilization, leukemia, and solid tumors. Clarifying the CXCR4/CXCL12 axis and role of AMD3100 will help remove malignant cells from the bone marrow niche, rendering them more accessible to targeted therapeutic agents.
Collapse
Affiliation(s)
- Tao Liu
- Division of Hematology/Oncology, Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, 1760 Haygood Drive NE, HSRB E363, Atlanta, GA 30322 USA.,Department of Oncology, The Affiliated Jiangyin Hospital of Southeast University Medical College, Wuxi, 214400 Jiangsu People's Republic of China
| | - Xiaobo Li
- Division of Hematology/Oncology, Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, 1760 Haygood Drive NE, HSRB E363, Atlanta, GA 30322 USA.,Tianjin Key Laboratory of Molecular Design and Drug Discovery, Tianjin Institute of Pharmaceutical Research, Tianjin, 300193 China
| | - Shuo You
- Department of Neurosurgery, Winship Cancer Institute, Emory University, Atlanta, GA 30322 USA
| | - Soumitra S Bhuyan
- School of Public Health, Division of Health Systems, Management, and Policy, The University of Memphis, Memphis, TN 38152 USA
| | - Lei Dong
- Division of Hematology/Oncology, Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, 1760 Haygood Drive NE, HSRB E363, Atlanta, GA 30322 USA
| |
Collapse
|
34
|
Byrd JC, Woyach JA, Johnson AJ. Translating PI3K-Delta Inhibitors to the Clinic in Chronic Lymphocytic Leukemia: The Story of CAL-101 (GS1101). Am Soc Clin Oncol Educ Book 2016:691-4. [PMID: 24451820 DOI: 10.14694/edbook_am.2012.32.75] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Targeted therapy with imatinib has transformed the treatment of chronic myeloid leukemia (CML). Unlike CML, chronic lymphocytic leukemia (CLL) lacks a common genetic aberration but does demonstrate constitutive activation of PI3-kinase (PI3K) as compared to normal B cells. This constitutively active PI3K in CLL likely relates to tonic B-cell receptor signaling that is present across a wide variety of B-cell malignancies. Although PI3K is quite proximal and represents an ideal target to pharmacologically modulate, the complexity of this pathway on which many normal functions are dependent had for many years been problematic. The p110 delta isoform of PI3K is relatively specific to hematopoietic cells, and elegant mouse studies where p110 delta was genetically inactivated demonstrated only a selective B-cell defect. Subsequent development of a potent, selective p110 delta inhibitor prompted translation into the clinic for the treatment of CLL and low-grade non-Hodgkin lymphoma (NHL). From the first patient treated where a dramatic early nodal response was noted, considerable excitement has developed for this class of drugs in CLL and NHL. We will summarize the development process of CAL-101 (now GS1101) in the treatment of chronic lymphoid malignancies such as CLL.
Collapse
Affiliation(s)
- John C Byrd
- From the Department of Internal Medicine, Division of Hematology, the Comprehensive Cancer Center at The Ohio State University, Columbus, OH; Division of Medicinal Chemistry, College of Pharmacy, and the Comprehensive Cancer Center at The Ohio State University, Columbus, OH
| | - Jennifer A Woyach
- From the Department of Internal Medicine, Division of Hematology, the Comprehensive Cancer Center at The Ohio State University, Columbus, OH; Division of Medicinal Chemistry, College of Pharmacy, and the Comprehensive Cancer Center at The Ohio State University, Columbus, OH
| | - Amy J Johnson
- From the Department of Internal Medicine, Division of Hematology, the Comprehensive Cancer Center at The Ohio State University, Columbus, OH; Division of Medicinal Chemistry, College of Pharmacy, and the Comprehensive Cancer Center at The Ohio State University, Columbus, OH
| |
Collapse
|
35
|
Mego M, Cholujova D, Minarik G, Sedlackova T, Gronesova P, Karaba M, Benca J, Cingelova S, Cierna Z, Manasova D, Pindak D, Sufliarsky J, Cristofanilli M, Reuben JM, Mardiak J. CXCR4-SDF-1 interaction potentially mediates trafficking of circulating tumor cells in primary breast cancer. BMC Cancer 2016; 16:127. [PMID: 26896000 PMCID: PMC4759765 DOI: 10.1186/s12885-016-2143-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 02/08/2016] [Indexed: 11/14/2022] Open
Abstract
Background Cytokines are involved in cancer invasion and metastasis. Circulating tumor cells (CTCs) play key role in tumor dissemination and are an independent survival predictor in breast cancer patients. The aim of this study was to assess correlation between CTCs and plasma cytokines in primary breast cancer (PBC) patients. Methods This study included 147 chemotherapy naïve PBC patients. Peripheral blood mononuclear cells (PBMC) were depleted of hematopoetic cells using RossetteSep™ negative selection kit. RNA extracted from CD45-depleted PBMC was interrogated for expression of EMT (Twist1, Snail1, Slug, Zeb1) and epithelial (Ck19) gene transcripts by qRT-PCR. The concentrations of 51 plasma cytokines were measured using multiplex bead arrays. Results CTCs were detected in 25.2 % patients. CTCs exhibiting only epithelial markers (CTC_EP) and only EMT markers (CTC_EMT) were present evenly in 11.6 % patients, while CTCs co-expressing both markers were detected in 2.0 % patients. Patients with presence of CTC_EP in peripheral blood had significantly elevated levels of plasma IFN-α2, IL-3, MCP-3, β-NGF, SCF, SCGF-β, TNF-β and SDF-1 compared to patients without CTC_EP. CTC_EP exhibited overexpression of SDF-1 receptor and CXCR4, but not other corresponding cytokine receptor, and in multivariate analysis SDF-1 was independently associated with CTC_EP. There was an inverse correlation between CTC_EMT and plasma cytokines CTACK, β-NGF and TRAIL, while presence of either subtype of CTCs was associated with increased level of TGF-β2. Conclusion Using cytokine profiling, we identified cytokines associated with CTCs subpopulations in peripheral blood of PBC. Our data suggest that CXCR4-SDF-1 axis is involved in mobilization and trafficking of epithelial CTCs.
Collapse
Affiliation(s)
- M Mego
- 2nd Department of Oncology, Faculty of Medicine, Comenius University, Klenova 1, 833 10, Bratislava, Slovak Republic. .,Translational Research Unit, Faculty of Medicine, Comenius University, Bratislava, Slovakia. .,National Cancer Institute, Bratislava, Slovakia.
| | - D Cholujova
- National Cancer Institute, Bratislava, Slovakia.
| | - G Minarik
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia.
| | - T Sedlackova
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia.
| | - P Gronesova
- Cancer Research Institute, Slovak Academy of Sciences, Slovak Medical University, Bratislava, Slovakia.
| | - M Karaba
- National Cancer Institute, Bratislava, Slovakia.
| | - J Benca
- National Cancer Institute, Bratislava, Slovakia.
| | - S Cingelova
- National Cancer Institute, Bratislava, Slovakia.
| | - Z Cierna
- Department of Pathology, Faculty of Medicine, Comenius University, Bratislava, Slovakia.
| | - D Manasova
- Translational Research Unit, Faculty of Medicine, Comenius University, Bratislava, Slovakia. .,National Cancer Institute, Bratislava, Slovakia.
| | - D Pindak
- National Cancer Institute, Bratislava, Slovakia. .,Slovak Medical University, Bratislava, Slovakia.
| | - J Sufliarsky
- 2nd Department of Oncology, Faculty of Medicine, Comenius University, Klenova 1, 833 10, Bratislava, Slovak Republic. .,National Cancer Institute, Bratislava, Slovakia.
| | - M Cristofanilli
- Robert H Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA.
| | - J M Reuben
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center Houston, Houston, TX, USA.
| | - J Mardiak
- 2nd Department of Oncology, Faculty of Medicine, Comenius University, Klenova 1, 833 10, Bratislava, Slovak Republic. .,National Cancer Institute, Bratislava, Slovakia.
| |
Collapse
|
36
|
Westhoff MA, Marschall N, Debatin KM. Novel Approaches to Apoptosis-Inducing Therapies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 930:173-204. [PMID: 27558822 DOI: 10.1007/978-3-319-39406-0_8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Induction of apoptotic programmed cell death is one of the underlying principles of most current cancer therapies. In this review, we discuss the limitations and drawbacks of this approach and identify three distinct, but overlapping strategies to avoid these difficulties and further enhance the efficacy of apoptosis-inducing therapies. We postulate that the application of multi-targeted small molecule inhibitor cocktails will reduce the risk of the cancer cell populations developing resistance towards therapy. Following from these considerations regarding population genetics and ecology, we advocate the reconsideration of therapeutic end points to maximise the benefits, in terms of quantity and quality of life, for the patients. Finally, combining both previous points, we also suggest an altered focus on the cellular and molecular targets of therapy, i.e. targeting the (cancer cells') interaction with the tumour microenvironment.
Collapse
Affiliation(s)
- Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Eythstrasse 24, 89075, Ulm, Germany
| | - Nicolas Marschall
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Eythstrasse 24, 89075, Ulm, Germany
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Eythstrasse 24, 89075, Ulm, Germany.
| |
Collapse
|
37
|
Abstract
In the past several years we have seen the identification and validation of several key pathways that drive malignant B-cell development. In addition, the effect nonmalignant effector cells within the immune microenvironment have on tumor survival, proliferation, and possibly chemotherapy resistance is increasingly understood. Although there is still much to be learned, this improved understanding combined with rapid advances in medicinal chemistry focusing on structure-based drug design have resulted in a shift in the development of new agents away from traditional chemotherapy to more selective agents targeting key cellular pathways. Examples of "hot" new therapeutic targets include the B-cell receptor signaling pathway, PI3K/mTOR/AKT pathway, histone deacetylases (HDAC), regulators of apoptosis such as the BCL-2 family, the proteasome, and cell-cell interactions within the tumor environment. Many drugs that target specific agents in early clinical development have demonstrated activity in various subtypes of lymphoma and leukemia. Monoclonal and conjugated antibodies targeting cell surface proteins such as CD19, CD22, CD37, and different epitopes of CD20 have also shown promise in relapsed B-cell malignancies and are rapidly moving into efficacy studies. This review will focus on a few of the new nonantibody-based targeted agents in development, their respective pathways, and their activity in various B-cell malignancies.
Collapse
Affiliation(s)
- Nathan Fowler
- From the Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX
| | | |
Collapse
|
38
|
Blunt MD, Steele AJ. Pharmacological targeting of PI3K isoforms as a therapeutic strategy in chronic lymphocytic leukaemia. Leuk Res Rep 2015; 4:60-3. [PMID: 26500849 PMCID: PMC4588368 DOI: 10.1016/j.lrr.2015.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 09/11/2015] [Indexed: 11/12/2022] Open
Abstract
PI3Kδ inhibitors such as idelalisib are providing improved therapeutic options for the treatment of chronic lymphocytic leukaemia (CLL). However under certain conditions, inhibition of a single PI3K isoform can be compensated by the other PI3K isoforms, therefore PI3K inhibitors which target multiple PI3K isoforms may provide greater efficacy. The development of compounds targeting multiple PI3K isoforms (α, β, δ, and γ) in CLL cells, in vitro, resulted in sustained inhibition of BCR signalling but with enhanced cytotoxicity and the potential for improve clinical responses. This review summarises the progress of PI3K inhibitor development and describes the rationale and potential for targeting multiple PI3K isoforms. Inhibition of PI3K may still be effective in CLL patients resistant to ibrutinib. Functional redundancy between PI3K isoforms may be a mechanism of drug resistance. Targeting multiple PI3K isoforms can increase cytotoxicity against CLL cells.
Collapse
Affiliation(s)
- Matthew D Blunt
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, SO16 6YD, United Kingdom
| | - Andrew J Steele
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, SO16 6YD, United Kingdom
| |
Collapse
|
39
|
Bermúdez Brito M, Goulielmaki E, Papakonstanti EA. Focus on PTEN Regulation. Front Oncol 2015; 5:166. [PMID: 26284192 PMCID: PMC4515857 DOI: 10.3389/fonc.2015.00166] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 07/07/2015] [Indexed: 12/17/2022] Open
Abstract
The role of phosphatase and tensin homolog on chromosome 10 (PTEN) as a tumor suppressor has been for a long time attributed to its lipid phosphatase activity against PI(3,4,5)P3, the phospholipid product of the class I PI3Ks. Besides its traditional role as a lipid phosphatase at the plasma membrane, a wealth of data has shown that PTEN can function independently of its phosphatase activity and that PTEN also exists and plays a role in the nucleus, in cytoplasmic organelles, and extracellularly. Accumulating evidence has shed light on diverse physiological functions of PTEN, which are accompanied by a complex regulation of its expression and activity. PTEN levels and function are regulated transcriptionally, post-transcriptionally, and post-translationally. PTEN is also sensitive to regulation by its interacting proteins and its localization. Herein, we summarize the current knowledge on mechanisms that regulate the expression and enzymatic activity of PTEN and its role in human diseases.
Collapse
Affiliation(s)
- Miriam Bermúdez Brito
- Department of Biochemistry, School of Medicine, University of Crete , Heraklion , Greece
| | - Evangelia Goulielmaki
- Department of Biochemistry, School of Medicine, University of Crete , Heraklion , Greece
| | | |
Collapse
|
40
|
The PI3K/mTOR inhibitor PF-04691502 induces apoptosis and inhibits microenvironmental signaling in CLL and the Eµ-TCL1 mouse model. Blood 2015; 125:4032-41. [DOI: 10.1182/blood-2014-11-610329] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 05/02/2015] [Indexed: 01/17/2023] Open
Abstract
Key Points
PF-04691502 induces potent apoptosis in CLL cells and suppresses prosurvival anti–immunoglobulin M signaling and CXCL12-induced migration. PF-04691502 displays powerful antitumor effects in vivo in the Eμ-TCL1 mouse model.
Collapse
|
41
|
Göckeritz E, Kerwien S, Baumann M, Wigger M, Vondey V, Neumann L, Landwehr T, Wendtner CM, Klein C, Liu N, Hallek M, Frenzel LP, Krause G. Efficacy of phosphatidylinositol-3 kinase inhibitors with diverse isoform selectivity profiles for inhibiting the survival of chronic lymphocytic leukemia cells. Int J Cancer 2015; 137:2234-42. [DOI: 10.1002/ijc.29579] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 03/13/2015] [Indexed: 01/08/2023]
Affiliation(s)
- Elisa Göckeritz
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases University of Cologne; Cologne Germany
| | - Susan Kerwien
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
| | - Michael Baumann
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
| | - Marion Wigger
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
| | - Verena Vondey
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases University of Cologne; Cologne Germany
| | - Lars Neumann
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases University of Cologne; Cologne Germany
| | - Thomas Landwehr
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
| | - Clemens M. Wendtner
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
| | - Christian Klein
- Roche Pharmaceutical Research & Early Development, Roche Innovation Center Zurich; Switzerland
| | - Ningshu Liu
- Bayer HealthCare Pharmaceuticals, Global Drug Discovery, TRG Oncology; Berlin Germany
| | - Michael Hallek
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases University of Cologne; Cologne Germany
| | - Lukas P. Frenzel
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases University of Cologne; Cologne Germany
| | - Günter Krause
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn; Cologne Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases University of Cologne; Cologne Germany
| |
Collapse
|
42
|
Guo F, Wang Y, Liu J, Mok SC, Xue F, Zhang W. CXCL12/CXCR4: a symbiotic bridge linking cancer cells and their stromal neighbors in oncogenic communication networks. Oncogene 2015; 35:816-26. [DOI: 10.1038/onc.2015.139] [Citation(s) in RCA: 254] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 03/28/2015] [Accepted: 03/30/2015] [Indexed: 02/07/2023]
|
43
|
Kupsa T, Horacek JM, Jebavy L. The role of adhesion molecules in acute myeloid leukemia and (hemato)oncology: A systematic review. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2015; 159:1-11. [DOI: 10.5507/bp.2014.049] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 09/16/2014] [Indexed: 12/18/2022] Open
|
44
|
Significantly inhibitory effects of low molecular weight heparin (Fraxiparine) on the motility of lung cancer cells and its related mechanism. Tumour Biol 2015; 36:4689-97. [PMID: 25619477 DOI: 10.1007/s13277-015-3117-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 01/14/2015] [Indexed: 02/05/2023] Open
Abstract
Low molecular weight heparin (LMWH) improving the cancer survival has been attracting attention for many years. Our previous study found that LMWH (Fraxiparine) strongly downregulated the invasive, migratory, and adhesive ability of human lung adenocarcinoma A549 cells. Here, we aimed to further identify the antitumor effects and possible mechanisms of Fraxiparine on A549 cells and human highly metastatic lung cancer 95D cells. The ability of cell invasion, migration, and adhesion were measured by Transwell, Millicell, and MTT assays. FITC-labeled phalloidin was used to detect F-actin bundles in cells. Chemotactic migration was analyzed in a modified Transwell assay. Measurement of protein expression and phosphorylation activity of PI3K, Akt, and mTOR was performed with Western blot. Our studies found that Fraxiparine significantly inhibited the invasive, migratory, and adhesive characteristics of A549 and 95D cells after 24 h incubation and showed a dose-dependent manner. Fraxiparine influenced the actin cytoskeleton rearrangement of A549 and 95D cells by preventing F-actin polymerization. Moreover, Fraxiparine could significantly inhibit CXCL12-mediated chemotactic migration of A549 and 95D cells in a concentration-dependent manner. Furthermore, Fraxiparine might destroy the interaction between CXCL12-CXCR4 axis, then suppress the PI3K-Akt-mTOR signaling pathway in lung cancer cells. For the first time, our data indicated that Fraxiparine could significantly inhibit the motility of lung cancer cells by restraining the actin cytoskeleton reorganization, and its related mechanism might be through inhibiting PI3K-Akt-mTOR signaling pathway mediated by CXCL12-CXCR4 axis. Therefore, Fraxiparine would be a potential drug for lung cancer metastasis therapy.
Collapse
|
45
|
Bernard S, Danglade D, Gardano L, Laguillier C, Lazarian G, Roger C, Thieblemont C, Marzec J, Gribben J, Cymbalista F, Varin-Blank N, Ledoux D, Baran-Marszak F. Inhibitors of BCR signalling interrupt the survival signal mediated by the micro-environment in mantle cell lymphoma. Int J Cancer 2014; 136:2761-74. [DOI: 10.1002/ijc.29326] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 10/27/2014] [Indexed: 02/01/2023]
Affiliation(s)
- Sophie Bernard
- U978 Institut National de la Santé et de la Recherche Médicale; Bobigny France
- Labex Inflamex, Université Paris 13; Sorbonne Paris Cité Bobigny France
- Service d'Hématologie Biologique, Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris; Bobigny France
| | - Damien Danglade
- Service d'Hématologie Biologique, Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris; Bobigny France
| | - Laura Gardano
- U978 Institut National de la Santé et de la Recherche Médicale; Bobigny France
- Labex Inflamex, Université Paris 13; Sorbonne Paris Cité Bobigny France
| | - Christelle Laguillier
- Service de Biochimie, Hôpital Jean Verdier, Assistance Publique-Hôpitaux de Paris; Bondy France
| | - Gregory Lazarian
- Service d'Hématologie Biologique, Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris; Bobigny France
| | - Claudine Roger
- Service d'Hématologie Biologique, Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris; Bobigny France
| | - Catherine Thieblemont
- Hématologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris; Paris France
| | - Jacek Marzec
- Barts Cancer Institute, Queen Mary, University of London; London United Kingdom
| | - John Gribben
- Barts Cancer Institute, Queen Mary, University of London; London United Kingdom
| | - Florence Cymbalista
- U978 Institut National de la Santé et de la Recherche Médicale; Bobigny France
- Labex Inflamex, Université Paris 13; Sorbonne Paris Cité Bobigny France
- Service d'Hématologie Biologique, Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris; Bobigny France
| | - Nadine Varin-Blank
- U978 Institut National de la Santé et de la Recherche Médicale; Bobigny France
- Labex Inflamex, Université Paris 13; Sorbonne Paris Cité Bobigny France
| | - Dominique Ledoux
- U978 Institut National de la Santé et de la Recherche Médicale; Bobigny France
- Labex Inflamex, Université Paris 13; Sorbonne Paris Cité Bobigny France
| | - Fanny Baran-Marszak
- U978 Institut National de la Santé et de la Recherche Médicale; Bobigny France
- Labex Inflamex, Université Paris 13; Sorbonne Paris Cité Bobigny France
- Service d'Hématologie Biologique, Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris; Bobigny France
| |
Collapse
|
46
|
CHEN FANGYUAN, CAO LANFANG, WAN HAIXIA, ZHANG MINYUE, CAI JIAYI, SHEN LIJING, ZHONG JIHUA, ZHONG HUA. Quercetin enhances adriamycin cytotoxicity through induction of apoptosis and regulation of mitogen-activated protein kinase/extracellular signal-regulated kinase/c-Jun N-terminal kinase signaling in multidrug-resistant leukemia K562 cells. Mol Med Rep 2014; 11:341-8. [DOI: 10.3892/mmr.2014.2734] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Accepted: 09/29/2014] [Indexed: 11/05/2022] Open
|
47
|
Adekola KUA, Dalva Aydemir S, Ma S, Zhou Z, Rosen ST, Shanmugam M. Investigating and targeting chronic lymphocytic leukemia metabolism with the human immunodeficiency virus protease inhibitor ritonavir and metformin. Leuk Lymphoma 2014; 56:450-9. [PMID: 24828872 DOI: 10.3109/10428194.2014.922180] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Chronic lymphocytic leukemia (CLL) remains fatal due to the development of resistance to existing therapies. Targeting abnormal glucose metabolism sensitizes various cancer cells to chemotherapy and/or elicits toxicity. Examination of glucose dependency in CLL demonstrated variable sensitivity to glucose deprivation. Further evaluation of metabolic dependencies of CLL cells resistant to glucose deprivation revealed increased engagement of fatty acid oxidation upon glucose withdrawal. Investigation of glucose transporter expression in CLL reveals up-regulation of glucose transporter GLUT4. Treatment of CLL cells with human immunodeficiency (HIV) protease inhibitor ritonavir, which inhibits GLUT4, elicits toxicity similar to that elicited upon glucose deprivation. CLL cells resistant to ritonavir are sensitized by co-treatment with metformin, potentially targeting compensatory mitochondrial complex 1 activity. Ritonavir and metformin have been administered in humans for the treatment of diabetes in patients with HIV, demonstrating the tolerance to this combination in humans. Our studies strongly substantiate further investigation of Food and Drug Administration approved ritonavir and metformin for CLL.
Collapse
|
48
|
CXCL12/CXCR4 axis confers adriamycin resistance to human chronic myelogenous leukemia and oroxylin A improves the sensitivity of K562/ADM cells. Biochem Pharmacol 2014; 90:212-25. [PMID: 24858801 DOI: 10.1016/j.bcp.2014.05.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 05/11/2014] [Accepted: 05/13/2014] [Indexed: 12/14/2022]
Abstract
This study was aimed at investigating the reversal effect of oroxylin A, a naturally bioactive monoflavonoid separated and purified from Scutellaria baicalensis Georgi, in human chronic myeloid leukemia (CML) and the underlying mechanisms. The results showed that CXCL12 could enhance the resistance of K562 cells to adriamycin (ADM) by increasing the expression of CXCR4, up-regulating the downstream PI3K/Akt pathway, and promoting translocation of NF-κB dimers into nucleus and subsequently decreasing the expression of apoptosis-related proteins in K562 cells. And we found that ADM resistance was partially reversed by CXCR4 siRNA transfection. Moreover, the sensitivity enhancement of oroxylin A was demonstrated by decreasing the expression of CXCR4 at both protein and mRNA levels, via PI3K/Akt/NF-κB pathway and triggering the apoptosis pathway in vitro. In addition, the in vivo study showed that oroxylin A increased apoptosis of leukemic cells with low systemic toxicity, and the mechanism was the same as in vitro study. In conclusion, all these results showed that oroxylin A improved the sensitivity of K562/ADM cells by increasing apoptosis in leukemic cells and decreasing the expression of CXCR4 and PI3K/Akt/NF-κB pathway, and probably served as a most promising agent for CML treatment.
Collapse
|
49
|
Acosta YY, Montes-Casado M, Aragoneses-Fenoll L, Dianzani U, Portoles P, Rojo JM. Suppression of CD4+ T lymphocyte activation in vitro and experimental encephalomyelitis in vivo by the phosphatidyl inositol 3-kinase inhibitor PIK-75. Int J Immunopathol Pharmacol 2014; 27:53-67. [PMID: 24674679 DOI: 10.1177/039463201402700108] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Class IA phosphatidyl inositol-3 kinases (PI3-K) are important targets in cancer therapy and are essential to immune responses, particularly through costimulation by CD28 and ICOS. Thus, small PI3-K inhibitors are likely candidates to immune intervention. PIK-75 is an efficient inhibitor of the PI3-K p110alpha catalytic subunits that suppresses tumor growth, and its effects on immune and autoimmune responses should be studied. Here, we describe the effect of PIK-75 on different immune parameters in vitro and in vivo. PIK-75 at concentrations commonly used in vitro (≥0.1 μM) inhibited T and B cell activation by Concanavalin A and LPS, respectively, and survival of non-stimulated spleen cells. In naive CD4+ T lymphocytes, PIK-75 induced apoptosis of resting or activated cells that was prevented by caspase inhibitors. At low nanomolar concentrations (≤10 nM), PIK-75 inhibited naive CD4+ T cell proliferation, and IL-2 and IFN-gamma production induced by anti-CD3 plus anti-CD28. In activated CD4+ T blasts costimulated by ICOS, PIK-75 (less than 10 nM) inhibited IFN-gamma, IL-17A, or IL-21 secretion. Furthermore, PIK-75 (20 mg/kg p.o.) suppressed clinical symptoms in ongoing experimental autoimmune encephalomyelitis (EAE) and inhibited MOG-specific responses in vitro. Thus, PIK-75 is an efficient suppressor of EAE, modulating lymphocyte function and survival.
Collapse
Affiliation(s)
- Y Y Acosta
- Department of Molecular and Cellular Medicine, Centre of Biological Investigation, CSIC, Madrid, Spain
| | - M Montes-Casado
- Unit of Cellular Immunology, National Centre of Microbiology, Institute of Health Carlos III, Majadahonda, Madrid, Spain
| | - L Aragoneses-Fenoll
- Unit of Cellular Immunology, National Centre of Microbiology, Institute of Health Carlos III, Majadahonda, Madrid, Spain
| | - U Dianzani
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD) and Department of Health Sciences, A. Avogadro University of Eastern Piedmont, Novara, Italy
| | - P Portoles
- Unit of Cellular Immunology, National Centre of Microbiology, Institute of Health Carlos III, Majadahonda, Madrid, Spain
| | - J M Rojo
- Department of Molecular and Cellular Medicine, Centre of Biological Investigation, CSIC, Madrid, Spain
| |
Collapse
|
50
|
Matas-Céspedes A, Rodriguez V, Kalko SG, Vidal-Crespo A, Rosich L, Casserras T, Balsas P, Villamor N, Giné E, Campo E, Roué G, López-Guillermo A, Colomer D, Pérez-Galán P. Disruption of follicular dendritic cells-follicular lymphoma cross-talk by the pan-PI3K inhibitor BKM120 (Buparlisib). Clin Cancer Res 2014; 20:3458-71. [PMID: 24799524 DOI: 10.1158/1078-0432.ccr-14-0154] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To uncover the signaling pathways underlying follicular lymphoma-follicular dendritic cells (FL-FDC) cross-talk and its validation as new targets for therapy. EXPERIMENTAL DESIGN FL primary cells and cell lines were cocultured in the presence or absence of FDC. After 24 and 48 hours, RNA was isolated from FL cells and subjected to gene expression profiling (GEP) and data meta-analysis using DAVID and GSEA softwares. Blockade of PI3K pathway by the pan-PI3K inhibitor BKM120 (buparlisib; Novartis Pharmaceutical Corporation) and the effect of PI3K inhibition on FL-FDC cross-talk were analyzed by means of ELISA, RT-PCR, human umbilical vein endothelial cell tube formation, adhesion and migration assays, Western blot, and in vivo studies in mouse FL xenografts. RESULTS GEP of FL-FDC cocultures yields a marked modulation of FL transcriptome by FDC. Pathway assignment by DAVID and GSEA software uncovered an overrepresentation of genes related to angiogenesis, cell adhesion, migration, and serum-response factors. We demonstrate that the addition of the pan-PI3K inhibitor BKM120 to the cocultures was able to downregulate the expression and secretion of proangiogenic factors derived from FL-FDC cocultures, reducing in vitro and in vivo angiogenesis. Moreover, BKM120 efficiently counteracts FDC-mediated cell adhesion and impedes signaling and migration induced by the chemokine CXCL12. BKM120 inhibits both constitutive PI3K/AKT pathway and FDC- or CXCL12-induced PI3K/AKT pathway, hampers FDC survival signaling, and reduces cell proliferation of FL cells in vitro and in mouse xenografts. CONCLUSIONS These data support the use of BKM120 in FL therapy to counteract microenvironment-related survival signaling in FL cells.
Collapse
Affiliation(s)
- Alba Matas-Céspedes
- Authors' Affiliations: Experimental Therapeutics in Lymphoid Malignancies Group, Department of Hemato-Oncology
| | - Vanina Rodriguez
- Authors' Affiliations: Experimental Therapeutics in Lymphoid Malignancies Group, Department of Hemato-Oncology
| | | | - Anna Vidal-Crespo
- Authors' Affiliations: Experimental Therapeutics in Lymphoid Malignancies Group, Department of Hemato-Oncology
| | - Laia Rosich
- Authors' Affiliations: Experimental Therapeutics in Lymphoid Malignancies Group, Department of Hemato-Oncology
| | | | - Patricia Balsas
- Authors' Affiliations: Experimental Therapeutics in Lymphoid Malignancies Group, Department of Hemato-Oncology
| | | | - Eva Giné
- Department of Hematology, Hospital Clínic, IDIBAPS, Barcelona, Spain
| | - Elías Campo
- Hematopathology Unit, Department of Pathology; and
| | - Gaël Roué
- Authors' Affiliations: Experimental Therapeutics in Lymphoid Malignancies Group, Department of Hemato-Oncology
| | | | - Dolors Colomer
- Authors' Affiliations: Experimental Therapeutics in Lymphoid Malignancies Group, Department of Hemato-Oncology; Hematopathology Unit, Department of Pathology; and
| | - Patricia Pérez-Galán
- Authors' Affiliations: Experimental Therapeutics in Lymphoid Malignancies Group, Department of Hemato-Oncology;
| |
Collapse
|