1
|
Shpakov AO. Allosteric Regulation of G-Protein-Coupled Receptors: From Diversity of Molecular Mechanisms to Multiple Allosteric Sites and Their Ligands. Int J Mol Sci 2023; 24:6187. [PMID: 37047169 PMCID: PMC10094638 DOI: 10.3390/ijms24076187] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Allosteric regulation is critical for the functioning of G protein-coupled receptors (GPCRs) and their signaling pathways. Endogenous allosteric regulators of GPCRs are simple ions, various biomolecules, and protein components of GPCR signaling (G proteins and β-arrestins). The stability and functional activity of GPCR complexes is also due to multicenter allosteric interactions between protomers. The complexity of allosteric effects caused by numerous regulators differing in structure, availability, and mechanisms of action predetermines the multiplicity and different topology of allosteric sites in GPCRs. These sites can be localized in extracellular loops; inside the transmembrane tunnel and in its upper and lower vestibules; in cytoplasmic loops; and on the outer, membrane-contacting surface of the transmembrane domain. They are involved in the regulation of basal and orthosteric agonist-stimulated receptor activity, biased agonism, GPCR-complex formation, and endocytosis. They are targets for a large number of synthetic allosteric regulators and modulators, including those constructed using molecular docking. The review is devoted to the principles and mechanisms of GPCRs allosteric regulation, the multiplicity of allosteric sites and their topology, and the endogenous and synthetic allosteric regulators, including autoantibodies and pepducins. The allosteric regulation of chemokine receptors, proteinase-activated receptors, thyroid-stimulating and luteinizing hormone receptors, and beta-adrenergic receptors are described in more detail.
Collapse
Affiliation(s)
- Alexander O Shpakov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 194223 St. Petersburg, Russia
| |
Collapse
|
2
|
Guo F, Liu B, Li X, Wang H, Zhu X, Su Y, He C, Zhu M, Ding J, Xu Y, Zhao X, Wang Y, Shan R, Zhu J, Xie J, Ge Q, Fan L, Ding Y, Xie Y, Zhang C, Li H, Wang H, Zhou H. Mass balance, metabolic disposition, and pharmacokinetics of a novel selective inhibitor of PI3Kδ [ 14C] SHC014748M in healthy Chinese subjects following oral administration. Cancer Chemother Pharmacol 2023; 91:143-156. [PMID: 36572783 DOI: 10.1007/s00280-022-04493-5] [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: 05/18/2022] [Accepted: 11/15/2022] [Indexed: 12/28/2022]
Abstract
PURPOSE SHC014748M is a potent, novel selective PI3Kδ isoform inhibitor and is proposed for the treatment of non-Hodgkin lymphoma and chronic lymphocytic leukemia/small lymphocytic lymphoma. This study investigated the pharmacokinetics, mass balance, metabolism and excretion of SHC014748M in Chinese male subjects following a single oral dose of 150 mg (100 μCi) [14C] SHC014748M. METHODS Six healthy Chinese male subjects administrated an oral suspension of 150 mg (100 μCi) [14C] SHC014748M and the samples of blood, urine and feces were collected for measuring. Liquid chromatography-tandem mass spectrometry and liquid scintillation counter were utilized to obtain mass balance and the pharmacokinetic data. RESULTS The median Tmax for [14C]-radioactivity was 1.6 ± 0.5 h after the oral administration of [14C] SHC014748M and the mean Cmax was 3863 ± 354 ng Eq./mL in plasma, while the mean Cmax, t1/2 values and AUC0-∞ values for total radioactivity in whole blood were 2466 ± 518 ng Eq./mL, 32.2 ± 30.5 h and 66,236 ± 44,232 h * ng Eq./mL, respectively. Fecal excretion was proposed as the predominant elimination route, accounting for a mean of 90.68 ± 11.38% of the administered dose, whereas the mean urine excretion was 6.00 ± 1.48% within 336 h post-dose. The proposed major metabolic pathway of [14C] SHC014748M in the human body were as follows: (I) monooxidation, (II) glucuronide acid conjugation, and (III) monoxide-hydrogenation. CONCLUSIONS SHC014748M was absorbed, metabolized and excreted with unchanged SHC014748M as its main circulating component in plasma following oral administration. In addition, it was speculated that fecal excretion was the principal excretion pathway; meanwhile, monohydroxy, glucuronide conjugation, oxygen, and hydrogenation were the major clearance pathways of SHC014748M through urine and/or feces. TRIAL REGISTRATION The trial registration number: CTR20202505.
Collapse
Affiliation(s)
- Fei Guo
- National Institute of Clinical Drug Trials, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China.,Department of Radiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China.,Department of Medical Imaging Diagnosis, School of Medical Imaging, Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - Bingyan Liu
- National Institute of Clinical Drug Trials, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - Xiaoli Li
- National Institute of Clinical Drug Trials, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - Haidong Wang
- Department of Pharmacy, The Affiliated Lianyungang Hospital of Xuzhou Medical University/The First People's Hospital of Lianyungang, Lianyungang, 222000, People's Republic of China
| | - Xingyu Zhu
- National Institute of Clinical Drug Trials, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China.,School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - Yue Su
- National Institute of Clinical Drug Trials, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China.,School of Public Foundation, Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - Cuixia He
- National Institute of Clinical Drug Trials, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China.,School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - Minhui Zhu
- National Institute of Clinical Drug Trials, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China.,School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - Jiaxiang Ding
- National Institute of Clinical Drug Trials, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China.,School of Public Foundation, Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - Yuanyuan Xu
- National Institute of Clinical Drug Trials, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China.,School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - Xiangdi Zhao
- National Institute of Clinical Drug Trials, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China.,School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - Ying Wang
- National Institute of Clinical Drug Trials, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China.,School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - Rongfang Shan
- National Institute of Clinical Drug Trials, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China.,School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - Juan Zhu
- National Institute of Clinical Drug Trials, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - Jing Xie
- National Institute of Clinical Drug Trials, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - Qin Ge
- National Institute of Clinical Drug Trials, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - Ling Fan
- National Institute of Clinical Drug Trials, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - Yuzhou Ding
- National Institute of Clinical Drug Trials, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - Yunqiu Xie
- National Institute of Clinical Drug Trials, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - Chaoyang Zhang
- National Institute of Clinical Drug Trials, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - Hongtao Li
- National Institute of Clinical Drug Trials, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - Hongju Wang
- National Institute of Clinical Drug Trials, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China.
| | - Huan Zhou
- National Institute of Clinical Drug Trials, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China. .,School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, People's Republic of China. .,School of Public Foundation, Bengbu Medical College, Bengbu, Anhui, People's Republic of China.
| |
Collapse
|
3
|
Wu X, Qian L, Zhao H, Lei W, Liu Y, Xu X, Li J, Yang Z, Wang D, Zhang Y, Zhang Y, Tang R, Yang Y, Tian Y. CXCL12/CXCR4: An amazing challenge and opportunity in the fight against fibrosis. Ageing Res Rev 2023; 83:101809. [PMID: 36442720 DOI: 10.1016/j.arr.2022.101809] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/24/2022] [Accepted: 11/24/2022] [Indexed: 11/26/2022]
Abstract
Fibrosis is a pathological process caused by abnormal wound healing response, which often leads to excessive deposition of extracellular matrix, distortion of organ architecture, and loss of organ function. Aging is an important risk factor for the development of organ fibrosis. C-X-C receptor 4 (CXCR4) is the predominant chemokine receptor on fibrocytes, C-X-C motif ligand 12 (CXCL12) is the only ligand of CXCR4. Accumulated evidence have confirmed that CXCL12/CXCR4 can be involved in multiple pathological mechanisms in fibrosis, such as inflammation, immunity, epithelial-mesenchymal transition, and angiogenesis. In addition, CXCL12/CXCR4 have also been shown to improve fibrosis levels in many organs including the heart, liver, lung and kidney; thus, they are promising targets for anti-fibrotic therapy. Notably, inhibitors of CXCL12 or CXCR4 also play an important role in various fibrosis-related diseases. In summary, this review systematically summarizes the role of CXCL12/CXCR4 in fibrosis, and this information is of great significance for understanding CXCL12/CXCR4. This will also contribute to the design of further studies related to CXCL12/CXCR4 and fibrosis, and shed light on potential therapies for fibrosis.
Collapse
Affiliation(s)
- Xue Wu
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Lu Qian
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Huadong Zhao
- Department of General Surgery, Tangdu Hospital, The Airforce Medical University, Xi'an, China
| | - Wangrui Lei
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Yanqing Liu
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Xiaoling Xu
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Jiawen Li
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Zhi Yang
- Department of General Surgery, Tangdu Hospital, The Airforce Medical University, Xi'an, China
| | - Du Wang
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Yuchen Zhang
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Yan Zhang
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Ran Tang
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Yang Yang
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China.
| | - Ye Tian
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China.
| |
Collapse
|
4
|
López C, Burkhardt B, Chan JKC, Leoncini L, Mbulaiteye SM, Ogwang MD, Orem J, Rochford R, Roschewski M, Siebert R. Burkitt lymphoma. Nat Rev Dis Primers 2022; 8:78. [PMID: 36522349 DOI: 10.1038/s41572-022-00404-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/19/2022] [Indexed: 12/16/2022]
Abstract
Burkitt lymphoma (BL) is an aggressive form of B cell lymphoma that can affect children and adults. The study of BL led to the identification of the first recurrent chromosomal aberration in lymphoma, t(8;14)(q24;q32), and subsequent discovery of the central role of MYC and Epstein-Barr virus (EBV) in tumorigenesis. Most patients with BL are cured with chemotherapy but those with relapsed or refractory disease usually die of lymphoma. Historically, endemic BL, non-endemic sporadic BL and the immunodeficiency-associated BL have been recognized, but differentiation of these epidemiological variants is confounded by the frequency of EBV positivity. Subtyping into EBV+ and EBV- BL might better describe the biological heterogeneity of the disease. Phenotypically resembling germinal centre B cells, all types of BL are characterized by dysregulation of MYC due to enhancer activation via juxtaposition with one of the three immunoglobulin loci. Additional molecular changes commonly affect B cell receptor and sphingosine-1-phosphate signalling, proliferation, survival and SWI-SNF chromatin remodelling. BL is diagnosed on the basis of morphology and high expression of MYC. BL can be effectively treated in children and adolescents with short durations of high dose-intensity multiagent chemotherapy regimens. Adults are more susceptible to toxic effects but are effectively treated with chemotherapy, including modified versions of paediatric regimens. The outcomes in patients with BL are good in high-income countries with low mortality and few late effects, but in low-income and middle-income countries, BL is diagnosed late and is usually treated with less-effective regimens affecting the overall good outcomes in patients with this lymphoma.
Collapse
Affiliation(s)
- Cristina López
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany
| | - Birgit Burkhardt
- Non-Hodgkin's Lymphoma Berlin-Frankfurt-Münster (NHL-BFM) Study Center and Paediatric Hematology, Oncology and BMT, University Hospital Muenster, Muenster, Germany
| | - John K C Chan
- Department of Pathology, Queen Elizabeth Hospital, Hong Kong SAR, China
| | - Lorenzo Leoncini
- Section of Pathology, Department of Medical Biotechnology, University of Siena, Siena, Italy
| | - Sam M Mbulaiteye
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, USA
| | | | | | - Rosemary Rochford
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Mark Roschewski
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Reiner Siebert
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany.
| |
Collapse
|
5
|
Xu H, Tilley DG. Pepducin-mediated G Protein-Coupled Receptor Signaling in the Cardiovascular System. J Cardiovasc Pharmacol 2022; 80:378-385. [PMID: 35170495 PMCID: PMC9365886 DOI: 10.1097/fjc.0000000000001236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 01/29/2022] [Indexed: 01/31/2023]
Abstract
ABSTRACT Pepducins are small-lipidated peptides designed from the intracellular loops of G protein-coupled receptors (GPCRs) that act in an allosteric manner to modulate the activity of GPCRs. Over the past 2 decades, pepducins have progressed initially from pharmacologic tools used to manipulate GPCR activity in an orthosteric site-independent manner to compounds with therapeutic potential that have even been used safely in phase 1 and 2 clinical trials in human subjects. The effect of pepducins at their cognate receptors has been shown to vary between antagonist, partial agonist, and biased agonist outcomes in various primary and clonal cell systems, with even small changes in amino acid sequence altering these properties and their receptor selectivity. To date, pepducins designed from numerous GPCRs have been studied for their impact on pathologic conditions, including cardiovascular diseases such as thrombosis, myocardial infarction, and atherosclerosis. This review will focus in particular on pepducins designed from protease-activated receptors, C-X-C motif chemokine receptors, formyl peptide receptors, and the β2-adrenergic receptor. We will discuss the historic context of pepducin development for each receptor, as well as the structural, signaling, pathophysiologic consequences, and therapeutic potential for each pepducin class.
Collapse
Affiliation(s)
- Heli Xu
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | | |
Collapse
|
6
|
Barreca M, Spanò V, Raimondi MV, Bivacqua R, Giuffrida S, Montalbano A, Cavalli A, Bertoni F, Barraja P. GPCR Inhibition in Treating Lymphoma. ACS Med Chem Lett 2022; 13:358-364. [PMID: 38239337 PMCID: PMC10796172 DOI: 10.1021/acsmedchemlett.1c00600] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are important classes of cell surface receptors involved in multiple physiological functions. Aberrant expression, upregulation, and mutation of GPCR signaling pathways are frequent in many types of cancers, promoting hyperproliferation, angiogenesis, and metastasis. Recent studies showed that alterations of GPCRs are involved in different lymphoma types. Herein, we review the synthetic strategies to obtain GPCR inhibitors, focusing on CXCR4 inhibitors which represent most of the GPCR inhibitors available in the market or under preclinical investigations for these diseases.
Collapse
Affiliation(s)
- Marilia Barreca
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Virginia Spanò
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Maria V Raimondi
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Roberta Bivacqua
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Stefano Giuffrida
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Alessandra Montalbano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Andrea Cavalli
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, USI, Via Francesco Chiesa 5, 6500 Bellinzona, Switzerland
- Swiss Institute of Bioinformatics, Quartier Sorge - Batiment Amphipole, 1015 Lausanne, Switzerland
| | - Francesco Bertoni
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Via Francesco Chiesa 5, 6500 Bellinzona, Switzerland
- Oncology Institute of Southern Switzerland, Via Vincenzo Vela 6, 6500 Bellinzona, Switzerland
| | - Paola Barraja
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| |
Collapse
|
7
|
PET imaging: back in the game for gastric EMZL? Blood 2022; 139:154-155. [PMID: 35024811 DOI: 10.1182/blood.2021013964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 09/27/2021] [Indexed: 11/20/2022] Open
|
8
|
Olson KM, Traynor JR, Alt A. Allosteric Modulator Leads Hiding in Plain Site: Developing Peptide and Peptidomimetics as GPCR Allosteric Modulators. Front Chem 2021; 9:671483. [PMID: 34692635 PMCID: PMC8529114 DOI: 10.3389/fchem.2021.671483] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 08/02/2021] [Indexed: 12/17/2022] Open
Abstract
Allosteric modulators (AMs) of G-protein coupled receptors (GPCRs) are desirable drug targets because they can produce fewer on-target side effects, improved selectivity, and better biological specificity (e.g., biased signaling or probe dependence) than orthosteric drugs. An underappreciated source for identifying AM leads are peptides and proteins-many of which were evolutionarily selected as AMs-derived from endogenous protein-protein interactions (e.g., transducer/accessory proteins), intramolecular receptor contacts (e.g., pepducins or extracellular domains), endogenous peptides, and exogenous libraries (e.g., nanobodies or conotoxins). Peptides offer distinct advantages over small molecules, including high affinity, good tolerability, and good bioactivity, and specific disadvantages, including relatively poor metabolic stability and bioavailability. Peptidomimetics are molecules that combine the advantages of both peptides and small molecules by mimicking the peptide's chemical features responsible for bioactivity while improving its druggability. This review 1) discusses sources and strategies to identify peptide/peptidomimetic AMs, 2) overviews strategies to convert a peptide lead into more drug-like "peptidomimetic," and 3) critically analyzes the advantages, disadvantages, and future directions of peptidomimetic AMs. While small molecules will and should play a vital role in AM drug discovery, peptidomimetics can complement and even exceed the advantages of small molecules, depending on the target, site, lead, and associated factors.
Collapse
Affiliation(s)
- Keith M. Olson
- Department of Pharmacology and Edward F Domino Research Center, University of Michigan, Ann Arbor, MI, United States
| | - John R. Traynor
- Department of Pharmacology and Edward F Domino Research Center, University of Michigan, Ann Arbor, MI, United States
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, United States
| | - Andrew Alt
- Department of Pharmacology and Edward F Domino Research Center, University of Michigan, Ann Arbor, MI, United States
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
9
|
Ortiz Zacarías NV, Bemelmans MP, Handel TM, de Visser KE, Heitman LH. Anticancer opportunities at every stage of chemokine function. Trends Pharmacol Sci 2021; 42:912-928. [PMID: 34521537 DOI: 10.1016/j.tips.2021.08.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 02/01/2023]
Abstract
The chemokine system, comprising 48 chemokines and 23 receptors, is critically involved in several hallmarks of cancer. Yet, despite extensive efforts from the pharmaceutical sector, only two drugs aimed at this system are currently approved for clinical use against cancer. To date, numerous pharmacological approaches have been developed to successfully intervene at different stages of chemokine function: (i) chemokine availability; (ii) chemokine-glycosaminoglycan binding; and (iii) chemokine receptor binding. Many of these strategies have been tested in preclinical cancer models, and some have advanced to clinical trials as potential anticancer therapies. Here we will review the strategies and growing pharmacological toolbox for manipulating the chemokine system in cancer, and address novel methods poised for future (pre)clinical testing.
Collapse
Affiliation(s)
- Natalia V Ortiz Zacarías
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands; Oncode Institute, Leiden University, Leiden, The Netherlands
| | - Martijn P Bemelmans
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Tracy M Handel
- University of California San Diego, Skaggs School of Pharmacy and Pharmaceutical Sciences, La Jolla, CA, USA
| | - Karin E de Visser
- Oncode Institute, Leiden University, Leiden, The Netherlands; Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands; Department of Immunology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Laura H Heitman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands; Oncode Institute, Leiden University, Leiden, The Netherlands.
| |
Collapse
|
10
|
Gao X, Qin S, Wu Y, Chu C, Jiang B, Johnson RH, Kuang D, Zhang J, Wang X, Mehta A, Tew KD, Leone GW, Yu XZ, Wang H. Nuclear PFKP promotes CXCR4-dependent infiltration by T cell acute lymphoblastic leukemia. J Clin Invest 2021; 131:e143119. [PMID: 34255748 DOI: 10.1172/jci143119] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 07/01/2021] [Indexed: 12/12/2022] Open
Abstract
PFKP (phosphofructokinase, platelet), the major isoform of PFK1 expressed in T cell acute lymphoblastic leukemia (T-ALL), is predominantly expressed in the cytoplasm to carry out its glycolytic function. Our study showed that PFKP is a nucleocytoplasmic shuttling protein with functional nuclear export and nuclear localization sequences (NLSs). Cyclin D3/CDK6 facilitated PFKP nuclear translocation by dimerization and by exposing the NLS of PFKP to induce the interaction between PFKP and importin 9. Nuclear PFKP stimulated the expression of C-X-C chemokine receptor type 4 (CXCR4), a chemokine receptor regulating leukemia homing/infiltration, to promote T-ALL cell invasion, which depended on the activity of c-Myc. In vivo experiments showed that nuclear PFKP promoted leukemia homing/infiltration into the bone marrow, spleen, and liver, which could be blocked with CXCR4 antagonists. Immunohistochemical staining of tissues from a clinically well-annotated cohort of T cell lymphoma/leukemia patients showed nuclear PFKP localization in invasive cancers, but not in nonmalignant T lymph node or reactive hyperplasia. The presence of nuclear PFKP in these specimens correlated with poor survival in patients with T cell malignancy, suggesting the potential utility of nuclear PFKP as a diagnostic marker.
Collapse
Affiliation(s)
- Xueliang Gao
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Shenghui Qin
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yongxia Wu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Chen Chu
- Department of Cancer Biology, Dana-Farber Cancer Institute and.,Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Baishan Jiang
- Department of Cancer Biology, Dana-Farber Cancer Institute and
| | - Roger H Johnson
- Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Dong Kuang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jie Zhang
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Xi Wang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Anand Mehta
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Kenneth D Tew
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Gustavo W Leone
- Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Xue-Zhong Yu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA.,Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Haizhen Wang
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina, USA.,Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
11
|
Eiger DS, Boldizsar N, Honeycutt CC, Gardner J, Rajagopal S. Biased agonism at chemokine receptors. Cell Signal 2020; 78:109862. [PMID: 33249087 DOI: 10.1016/j.cellsig.2020.109862] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/07/2020] [Accepted: 11/24/2020] [Indexed: 12/11/2022]
Abstract
In the human chemokine system, interactions between the approximately 50 known endogenous chemokine ligands and 20 known chemokine receptors (CKRs) regulate a wide range of cellular functions and biological processes including immune cell activation and homeostasis, development, angiogenesis, and neuromodulation. CKRs are a family of G protein-coupled receptors (GPCR), which represent the most common and versatile class of receptors in the human genome and the targets of approximately one third of all Food and Drug Administration-approved drugs. Chemokines and CKRs bind with significant promiscuity, as most CKRs can be activated by multiple chemokines and most chemokines can activate multiple CKRs. While these ligand-receptor interactions were previously regarded as redundant, it is now appreciated that many chemokine:CKR interactions display biased agonism, the phenomenon in which different ligands binding to the same receptor signal through different pathways with different efficacies, leading to distinct biological effects. Notably, these biased responses can be modulated through changes in ligand, receptor, and or the specific cellular context (system). In this review, we explore the biochemical mechanisms, functional consequences, and therapeutic potential of biased agonism in the chemokine system. An enhanced understanding of biased agonism in the chemokine system may prove transformative in the understanding of the mechanisms and consequences of biased signaling across all GPCR subtypes and aid in the development of biased pharmaceuticals with increased therapeutic efficacy and safer side effect profiles.
Collapse
Affiliation(s)
| | - Noelia Boldizsar
- Trinity College of Arts and Sciences, Duke University, Durham, NC 27710, USA.
| | | | - Julia Gardner
- Trinity College of Arts and Sciences, Duke University, Durham, NC 27710, USA.
| | - Sudarshan Rajagopal
- Department of Biochemistry, Duke University, Durham, NC 27710, USA; Department of Medicine, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
12
|
Li L, Chai Y, Wu C, Zhao L. Chemokine receptor CXCR4: An important player affecting the molecular-targeted drugs commonly used in hematological malignancies. Expert Rev Hematol 2020; 13:1387-1396. [PMID: 33170753 DOI: 10.1080/17474086.2020.1839885] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION A variety of molecular-targeted drugs have been widely used in hematological malignancies and have shown great advances. Nevertheless, as the use of drugs in clinical practice increases, the problem of relapse or of the disease being refractory to treatment is becoming apparent. This problem is closely related to the C-X-C chemokine receptor 4 (CXCR4). AREAS COVERED This review focuses mainly on the effect of CXCR4 on molecular-targeted drug resistance in hematological malignancies as well as the clinical efficacy of CXCR4 antagonists combined with molecular-targeted drugs. Relevant literatures published between 2006 and 2020 were searched using PubMed/Medline for this review. EXPERT OPINION Monoclonal antibodies and non-antibody molecular-targeted drugs provide new therapeutic approaches for B-lineage malignancies and leukemia, but the clinical activity of these drugs is affected by CXCR4. In general, high CXCR4 expression or mutation inhibits the effects of molecular-targeted drugs, but there are exceptions, and in studies of proteasome inhibitors bortezomib (Bz) in multiple myeloma (MM), low CXCR4 expression or loss of CXCR4 was associated with Bz resistance (BzR) and poor treatment outcomes. Given that CXCR4 is a critical mediator of molecular-targeted drug resistance, numerous studies have combined molecular-targeted drugs with CXCR4 antagonists, which synergistically enhance the anti-proliferative/pro-apoptotic effect of molecular-targeted drugs.
Collapse
Affiliation(s)
- Liangliang Li
- The First Clinical Medical College of Lanzhou University , Lanzhou, Gansu, China.,Department of Hematology, Lanzhou University Second Hospital , Lanzhou, Gansu, China
| | - Ye Chai
- Department of Hematology, Lanzhou University Second Hospital , Lanzhou, Gansu, China
| | - ChongYang Wu
- Department of Hematology, Lanzhou University Second Hospital , Lanzhou, Gansu, China
| | - Li Zhao
- Department of Central Laboratory, The First Hospital of Lanzhou University , Lanzhou, Gansu, China
| |
Collapse
|
13
|
Rehman A, Baloch NUA, Morrow JP, Pacher P, Haskó G. Targeting of G-protein coupled receptors in sepsis. Pharmacol Ther 2020; 211:107529. [PMID: 32197794 PMCID: PMC7388546 DOI: 10.1016/j.pharmthera.2020.107529] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 03/11/2020] [Accepted: 03/11/2020] [Indexed: 12/11/2022]
Abstract
The Third International Consensus Definitions (Sepsis-3) define sepsis as life-threatening multi-organ dysfunction caused by a dysregulated host response to infection. Sepsis can progress to septic shock-an even more lethal condition associated with profound circulatory, cellular and metabolic abnormalities. Septic shock remains a leading cause of death in intensive care units and carries a mortality of almost 25%. Despite significant advances in our understanding of the pathobiology of sepsis, therapeutic interventions have not translated into tangible differences in the overall outcome for patients. Clinical trials of antagonists of various pro-inflammatory mediators in sepsis have been largely unsuccessful in the past. Given the diverse physiologic roles played by G-protein coupled receptors (GPCR), modulation of GPCR signaling for the treatment of sepsis has also been explored. Traditional pharmacologic approaches have mainly focused on ligands targeting the extracellular domains of GPCR. However, novel techniques aimed at modulating GPCR intracellularly through aptamers, pepducins and intrabodies have opened a fresh avenue of therapeutic possibilities. In this review, we summarize the diverse roles played by various subfamilies of GPCR in the pathogenesis of sepsis and identify potential targets for pharmacotherapy through these novel approaches.
Collapse
Affiliation(s)
- Abdul Rehman
- Department of Medicine, Rutgers-New Jersey Medical School, Newark, NJ, United States
| | - Noor Ul-Ain Baloch
- Department of Medicine, Rutgers-New Jersey Medical School, Newark, NJ, United States
| | - John P Morrow
- Department of Medicine, Columbia University, New York City, NY, United States
| | - Pál Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - György Haskó
- Department of Anesthesiology, Columbia University, New York City, NY, United States.
| |
Collapse
|
14
|
Adlere I, Caspar B, Arimont M, Dekkers S, Visser K, Stuijt J, de Graaf C, Stocks M, Kellam B, Briddon S, Wijtmans M, de Esch I, Hill S, Leurs R. Modulators of CXCR4 and CXCR7/ACKR3 Function. Mol Pharmacol 2019; 96:737-752. [DOI: 10.1124/mol.119.117663] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 09/14/2019] [Indexed: 02/06/2023] Open
|
15
|
Chen L, Ouyang J, Wienand K, Bojarczuk K, Hao Y, Chapuy B, Neuberg D, Juszczynski P, Lawton LN, Rodig SJ, Monti S, Shipp MA. CXCR4 upregulation is an indicator of sensitivity to B-cell receptor/PI3K blockade and a potential resistance mechanism in B-cell receptor-dependent diffuse large B-cell lymphomas. Haematologica 2019; 105:1361-1368. [PMID: 31471373 PMCID: PMC7193488 DOI: 10.3324/haematol.2019.216218] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 09/26/2019] [Indexed: 12/30/2022] Open
Abstract
B-cell receptor (BCR) signaling pathway components represent promising treatment targets in multiple B-cell malignancies including diffuse large B-cell lymphoma (DLBCL). In in vitro and in vivo model systems, a subset of DLBCLs depend upon BCR survival signals and respond to proximal BCR/phosphoinositide 3 kinase (PI3K) blockade. However, single-agent BCR pathway inhibitors have had more limited activity in patients with DLBCL, underscoring the need for indicators of sensitivity to BCR blockade and insights into potential resistance mechanisms. Here, we report highly significant transcriptional upregulation of C-X-C chemokine receptor 4 (CXCR4) in BCR-dependent DLBCL cell lines and primary tumors following chemical spleen tyrosine kinase (SYK) inhibition, molecular SYK depletion or chemical PI3K blockade. SYK or PI3K inhibition also selectively upregulated cell surface CXCR4 protein expression in BCR-dependent DLBCLs. CXCR4 expression was directly modulated by fork-head box O1 via the PI3K/protein kinase B/forkhead box O1 signaling axis. Following chemical SYK inhibition, all BCR-dependent DLBCLs exhibited significantly increased stromal cell-derived factor-1α (SDF-1α) induced chemotaxis, consistent with the role of CXCR4 signaling in B-cell migration. Select PI3K isoform inhibitors also augmented SDF-1α induced chemotaxis. These data define CXCR4 upregulation as an indicator of sensitivity to BCR/PI3K blockade and identify CXCR4 signaling as a potential resistance mechanism in BCR-dependent DLBCLs.
Collapse
Affiliation(s)
- Linfeng Chen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Current address: H3 Biomedicine, Cambridge, MA, USA
| | - Jing Ouyang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kirsty Wienand
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kamil Bojarczuk
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Current address: Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Yansheng Hao
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Current Address: Department of Pathology, Mount Sinai Hospital, New York, NY, USA
| | - Bjoern Chapuy
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Current Address: Department of Hematology and Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Donna Neuberg
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, USA
| | - Przemyslaw Juszczynski
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Current address: Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Lee N Lawton
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Scott J Rodig
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Stefano Monti
- Section of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Margaret A Shipp
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
16
|
Du H, Gao L, Luan J, Zhang H, Xiao T. C-X-C Chemokine Receptor 4 in Diffuse Large B Cell Lymphoma: Achievements and Challenges. Acta Haematol 2019; 142:64-70. [PMID: 31096215 DOI: 10.1159/000497430] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 02/02/2019] [Indexed: 12/24/2022]
Abstract
Diffuse large B cell lymphoma (DLBCL), an aggressive cancer of the B cells, is the most common subtype of non-Hodgkin lymphoma (NHL) worldwide. In China, the cases of DLBCL increase yearly. C-X-C chemokine receptor 4 (CXCR4) has been implicated in the migration and trafficking of malignant B cells in several hematological malignancies, and only a few reports have been published on the role of CXCR4 in the metastasis of DLBCL. This review summarizes the relevant perspectives on the functional mechanism, prognostic significance, and therapeutic applications of the CXCL12/CXCR4 axis in DLBCL, in particular DLBCL with bone marrow involvement.
Collapse
Affiliation(s)
- Hui Du
- Division of Hematology, Liaocheng People's Hospital, Liaocheng, China,
| | - Lei Gao
- Division of Hematology, Liaocheng People's Hospital, Liaocheng, China
| | - Jing Luan
- Division of Hematology, Liaocheng People's Hospital, Liaocheng, China
| | - Hangfan Zhang
- Division of Hematology, Liaocheng People's Hospital, Liaocheng, China
| | - Taiwu Xiao
- Division of Hematology, Liaocheng People's Hospital, Liaocheng, China
| |
Collapse
|
17
|
Abstract
PURPOSE OF REVIEW In addition to the recent progresses in the description of the genetic landscape of B-cell non-Hodgkin's lymphomas, tumor microenvironment has progressively emerged as a central determinant of early lymphomagenesis, subclonal evolution, drug resistance, and late progression/transformation. The purpose of this review is to outline the most recent findings regarding malignant B-cell niche composition and organization supporting direct and indirect tumor-promoting functions of lymphoma microenvironment. RECENT FINDINGS Lymphoma supportive niche integrates a dynamic and orchestrated network of immune and stromal cell subsets producing, with a high level of spatial and kinetic heterogeneity, extracellular and membrane factors regulating tumor migration, survival, proliferation, immune escape, as well as tumor microarchitecture, and mechanical constraints. Some recent insights have improved our understanding of these various components of lymphoma microenvironment, taking into account the mechanisms underlying the coevolution of malignant and nonmalignant cells within the tumor niche. SUMMARY Deciphering tumor niche characteristics, functions, and origin could offer new therapeutic opportunities through the targeting of pivotal cellular and molecular components of the supportive microenvironment, favoring immune cell reactivation and infiltration, and/or limiting tumor retention within this protective niche.
Collapse
|
18
|
Laursen MB, Reinholdt L, Schönherz AA, Due H, Jespersen DS, Grubach L, Ettrup MS, Røge R, Falgreen S, Sørensen S, Bødker JS, Schmitz A, Johnsen HE, Bøgsted M, Dybkær K. High CXCR4 expression impairs rituximab response and the prognosis of R-CHOP-treated diffuse large B-cell lymphoma patients. Oncotarget 2019; 10:717-731. [PMID: 30774774 PMCID: PMC6366826 DOI: 10.18632/oncotarget.26588] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 12/29/2018] [Indexed: 12/22/2022] Open
Abstract
Survival of diffuse large B-cell lymphoma (DLBCL) patients has improved by inclusion of rituximab. Refractory/recurrent disease caused by treatment resistance is, however, a major problem. Determinants of rituximab sensitivity are not fully understood, but effect of rituximab are enhanced by antagonizing cell surface receptor CXCR4. In a two-step strategy, we tested the hypothesis that prognostic value of CXCR4 in DLBCL relates to rituximab treatment, due to a hampering effect of CXCR4 on the response of DLBCL cells to rituximab. First, by investigating the prognostic impact of CXCR4 mRNA expression separately for CHOP (n=181) and R-CHOP (n=233) cohorts and, second, by assessing the interaction between CXCR4 and rituximab in DLBCL cell lines. High CXCR4 expression level was significantly associated with poor outcome only for R-CHOP-treated patients, independent of IPI score, CD20 expression, ABC/GCB and B-cell-associated gene signature (BAGS) classifications. s. For responsive cell lines, inverse correlation was observed between rituximab sensitivity and CXCR4 surface expression, rituximab induced upregulation of surface-expressed CXCR4, and growth-inhibitory effect of rituximab increased by plerixafor, supporting negative impact of CXCR4 on rituximab function. In conclusion, CXCR4 is a promising independent prognostic marker for R-CHOP-treated DLBCL patients, possibly due to inverse correlation between CXCR4 expression and rituximab sensitivity.
Collapse
Affiliation(s)
| | - Linn Reinholdt
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark
| | | | - Hanne Due
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark
| | | | - Lykke Grubach
- Department of Hematopathology, Aalborg University Hospital, Aalborg, Denmark
| | | | - Rasmus Røge
- Department of Hematopathology, Aalborg University Hospital, Aalborg, Denmark
| | - Steffen Falgreen
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark
| | - Suzette Sørensen
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark.,Centre for Clinical Research, North Denmark Regional Hospital, Hjørring, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Julie Støve Bødker
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark.,Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Alexander Schmitz
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark.,Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Hans E Johnsen
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark.,Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Martin Bøgsted
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark.,Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Karen Dybkær
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark.,Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
19
|
Du H, Zhang L, Li G, Liu W, Tang W, Zhang H, Luan J, Gao L, Wang X. CXCR4 and CCR7 Expression in Primary Nodal Diffuse Large B-Cell Lymphoma-A Clinical and Immunohistochemical Study. Am J Med Sci 2019; 357:302-310. [PMID: 30904045 DOI: 10.1016/j.amjms.2019.01.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 11/08/2018] [Accepted: 01/15/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND A few studies have evaluated the expression of chemokine receptors CXCR4 and CCR7 in diffuse large B-cell lymphoma (DLBCL); however, the association between CXCR4 and CCR7 with bone marrow (BM) involvement and their synergistic effect on prognosis is still unclear. Our study investigated this aspect. METHODS Specimens were obtained from 61 primary nodal DLBCL patients and 100 reactive proliferative lymphadenitis patients. CXCR4 and CCR7 expression levels were examined by immunohistochemical staining; the relationship between these levels and clinical parameters and the differences in overall survival were analyzed. RESULTS CXCR4 and CCR7 overexpression was observed in the malignant lymph node tissues from most DLBCL patients. CCR7 expression was significantly higher in the non-GCB than the GCB subtype; CXCR4 positivity rates showed no significant difference between the 2 subtypes. In DLBCL patients with BM involvement, CXCR4 was overexpressed in almost all BM samples, but CCR7 expression was low in BM. CXCR4 overexpression was associated with advanced Ann Arbor stages, MYC overexpression, and increased extranodal infiltration; CCR7 was associated with advanced Ann Arbor stages and elevated LDH. Like the case for CCR7, the survival rate of CXCR4-positive DLBCL patients was significantly lower than that of the CXCR4-negative patients. CXCR4+CCR7+ patients had the lowest survival rate. CONCLUSIONS There is a positive correlation between CXCR4 overexpression and BM involvement. CXCR4 and CCR7 overexpression is associated with poorer overall survival, especially in CXCR4 and CCR7 copositive patients. CXCR4, CCR7, Ki-67 index, and MYC were independent prognostic factors for DLBCL. Blocking CXCR4 and/or CCR7 can be a novel therapeutic strategy for DLBCL.
Collapse
Affiliation(s)
- Hui Du
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, People's Republic of China; Division of Hematology
| | | | | | - Wei Liu
- Central Laboratory, Liaocheng People's Hospital, Liaocheng, Shandong, People's Republic of China
| | - Wenqiang Tang
- Central Laboratory, Liaocheng People's Hospital, Liaocheng, Shandong, People's Republic of China
| | | | | | | | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, People's Republic of China.
| |
Collapse
|
20
|
Barr TP, Garzia C, Guha S, Fletcher EK, Nguyen N, Wieschhaus AJ, Ferrer L, Covic L, Kuliopulos A. PAR2 Pepducin-Based Suppression of Inflammation and Itch in Atopic Dermatitis Models. J Invest Dermatol 2018; 139:412-421. [PMID: 30287285 DOI: 10.1016/j.jid.2018.08.019] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 08/09/2018] [Accepted: 08/12/2018] [Indexed: 02/06/2023]
Abstract
PAR2 has been proposed to contribute to lesion formation and intense itch in atopic dermatitis. Here, we tested the ability of a cell-penetrating pepducin, PZ-235, to mitigate the potentially deleterious effects of PAR2 in models of atopic dermatitis. PZ-235 significantly inhibited PAR2-mediated expression of inflammatory factors NF-κB, TSLP, TNF-α, and differentiation marker K10 by 94%-98% (P < 0.001) in human keratinocytes and suppressed IL-4 and IL-13 by 68%-83% (P < 0.05) in mast cells. In delayed pepducin treatment models of oxazolone- and DNFB-induced dermatitis, PZ-235 significantly attenuated skin thickening by 43%-100% (P < 0.01) and leukocyte crusting by 57% (P < 0.05), and it inhibited ex vivo chemotaxis of leukocytes toward PAR2 agonists. Daily PZ-235 treatment of filaggrin-deficient mice exposed to dust mite allergens for 8 weeks significantly suppressed total leukocyte and T-cell infiltration by 50%-68%; epidermal thickness by 60%-77%; and skin thickening, scaling, excoriation, and total lesion severity score by 46%-56%. PZ-235 significantly reduced itching caused by wasp venom peptide degranulation of mast cells in mice by 51% (P < 0.05), which was comparable to the protective effects conferred by PAR2 deficiency. Taken together, these results suggest that a PAR2 pepducin may confer broad therapeutic benefits as a disease-modifying treatment for atopic dermatitis and itch.
Collapse
Affiliation(s)
- Travis P Barr
- Center of Hemostasis and Thrombosis Research, Tufts Medical Center, Department of Medicine, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Chris Garzia
- Center of Hemostasis and Thrombosis Research, Tufts Medical Center, Department of Medicine, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Srijoy Guha
- Center of Hemostasis and Thrombosis Research, Tufts Medical Center, Department of Medicine, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Elizabeth K Fletcher
- Center of Hemostasis and Thrombosis Research, Tufts Medical Center, Department of Medicine, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Nga Nguyen
- Center of Hemostasis and Thrombosis Research, Tufts Medical Center, Department of Medicine, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Adam J Wieschhaus
- Center of Hemostasis and Thrombosis Research, Tufts Medical Center, Department of Medicine, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Lluis Ferrer
- Department of Dermatology, Tufts Cummings School of Veterinary Medicine, North Grafton, Massachusetts, USA
| | - Lidija Covic
- Center of Hemostasis and Thrombosis Research, Tufts Medical Center, Department of Medicine, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Athan Kuliopulos
- Center of Hemostasis and Thrombosis Research, Tufts Medical Center, Department of Medicine, Tufts University School of Medicine, Boston, Massachusetts, USA.
| |
Collapse
|
21
|
Comparison of interleukin-2-inducible kinase (ITK) inhibitors and potential for combination therapies for T-cell lymphoma. Sci Rep 2018; 8:14216. [PMID: 30242208 PMCID: PMC6154993 DOI: 10.1038/s41598-018-32634-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 08/17/2018] [Indexed: 02/07/2023] Open
Abstract
Patients with peripheral T-cell lymphomas generally have poor clinical outcomes with conventional chemotherapy. Recent advances have demonstrated that a large subgroup of PTCL are derived from follicular helper (Tfh) T-cells. These cases show a characteristic pattern of gene expression, which includes high-level protein expression of interleukin-2-inducible kinase (ITK). ITK is a member of the TEC family of kinases and normally has essential functions in regulating T-cell receptor signalling and T-cell differentiation. Here we report a side-by-side comparison of four ITK inhibitors. We investigate effects on apoptosis, phosphorylation of signaling molecules, calcium flux and migration. In line with a specific mechanism of action ONO7790500 and BMS509744 did not inhibit MEK1/2 or AKT phosphorylation although other ITK inhibitors, ibrutinib and PF-06465469, did have this effect. Specific ITKi had modest effects on apoptosis alone but there was definite synergy with doxorubicin, pictilisib (PI3Ki) and idelalisib (PI3Kδi). ITKi repressed migration of Jurkat cells caused by CXCL12 and the CXCR4 antagonist, plerixafor enhanced this effect. Overall ITKi may have several mechanisms of action that will be therapeutically useful in PTCL including reduction in survival and perturbation of trafficking.
Collapse
|
22
|
Covic L, Kuliopulos A. Protease-Activated Receptor 1 as Therapeutic Target in Breast, Lung, and Ovarian Cancer: Pepducin Approach. Int J Mol Sci 2018; 19:ijms19082237. [PMID: 30065181 PMCID: PMC6121574 DOI: 10.3390/ijms19082237] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 07/19/2018] [Accepted: 07/25/2018] [Indexed: 12/20/2022] Open
Abstract
The G-protein coupled receptors (GPCRs) belong to a large family of diverse receptors that are well recognized as pharmacological targets. However, very few of these receptors have been pursued as oncology drug targets. The Protease-activated receptor 1 (PAR1), which is a G-protein coupled receptor, has been shown to act as an oncogene and is an emerging anti-cancer drug target. In this paper, we provide an overview of PAR1’s biased signaling role in metastatic cancers of the breast, lungs, and ovaries and describe the development of PAR1 inhibitors that are currently in clinical use to treat acute coronary syndromes. PAR1 inhibitor PZ-128 is in a Phase II clinical trial and is being developed to prevent ischemic and thrombotic complication of patients undergoing cardiac catheterization. PZ-128 belongs to a new class of cell-penetrating, membrane-tethered peptides named pepducins that are based on the intracellular loops of receptors targeting the receptor G-protein interface. Application of PZ-128 as an anti-metastatic and anti-angiogenic therapeutic agent in breast, lung, and ovarian cancer is being reviewed.
Collapse
Affiliation(s)
- Lidija Covic
- Division of Hematology/Oncology, Tufts Medical Center, Boston, MA 02111, USA.
- Department of Medicine, Tufts Medical Center, Boston, MA 02111, USA.
- Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Boston, MA 02111, USA.
| | - Athan Kuliopulos
- Division of Hematology/Oncology, Tufts Medical Center, Boston, MA 02111, USA.
- Department of Medicine, Tufts Medical Center, Boston, MA 02111, USA.
- Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Boston, MA 02111, USA.
| |
Collapse
|
23
|
Fouquet G, Guidez S, Richez V, Stoppa AM, Le Tourneau C, Macro M, Gruchet C, Bobin A, Moya N, Syshenko T, Sabirou F, Levy A, Franques P, Gardeney H, Karlin L, Benboubker L, Ouali M, Vedovato JC, Ferre P, Pavlyuk M, Attal M, Facon T, Leleu X. Phase I dose-escalation study of F50067, a humanized anti-CXCR4 monoclonal antibody alone and in combination with lenalidomide and low-dose dexamethasone, in relapsed or refractory multiple myeloma. Oncotarget 2018; 9:23890-23899. [PMID: 29844860 PMCID: PMC5963612 DOI: 10.18632/oncotarget.25156] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 03/29/2018] [Indexed: 11/25/2022] Open
Abstract
Purpose Multiple myeloma (MM) remains an incurable disease as tumor cells ultimately resist to all available drugs. Homing of tumor cells to the bone marrow microenvironment, involving especially the CXCR4/SDF-1 axis, allows them to survive, proliferate and resist to therapy. F50067, a humanized anti-CXCR4 IgG1 antibody, has promising preclinical activity in MM. We present a phase I multicenter escalation study in relapsed/refractory MM (RRMM) to determine the maximum tolerated dose (MTD) for F50067 alone and in combination with lenalidomide and low dose dexamethasone (Len-Dex). Experimental design 14 end-stage RRMM patients received F50067 single agent (n = 10) or in combination with Len-Dex (n = 4). Results One dose-limiting toxicity was observed, a grade 4 neutropenia lasting more than 7 days in combination arm. MTD could not be established. Thrombocytopenia was observed in 100% and neutropenia in 92.9% of patients with no cases of febrile neutropenia and no severe bleeding or hematoma. Non-hematological adverse events were of mild to moderate severity. Nine patients (6 in single arm and 3 in combination arm) were evaluable for response, with 66.7% overall response rate (≥PR) in combination arm, and 33.3% of disease control (≥SD) in single agent arm. At the time of study termination, 55.6% had progressed. Conclusion This study suggests that egression of tumor cells to the blood stream can represent a novel therapeutic strategy for MM. However, because of significant hematological toxicity, this study had to be discontinued. Further studies are needed to validate the feasibility of this approach in clinical practice.
Collapse
Affiliation(s)
- Guillemette Fouquet
- Institut Imagine, Unité Inserm U1163, Centre National de la Recherche Scientifique CNRS ERL8254, Paris, France
| | - Stéphanie Guidez
- Hôpital La Milétrie, Centre Hospitalier Universitaire, Poitiers, France.,Inserm Centre d'Investigation Clinique U1402, Centre Hospitalier Universitaire, Poitiers, France
| | - Valentine Richez
- Hôpital La Milétrie, Centre Hospitalier Universitaire, Poitiers, France
| | | | | | | | - Cécile Gruchet
- Hôpital La Milétrie, Centre Hospitalier Universitaire, Poitiers, France
| | - Arthur Bobin
- Hôpital La Milétrie, Centre Hospitalier Universitaire, Poitiers, France
| | - Niels Moya
- Hôpital La Milétrie, Centre Hospitalier Universitaire, Poitiers, France
| | - Thomas Syshenko
- Hôpital La Milétrie, Centre Hospitalier Universitaire, Poitiers, France
| | - Florence Sabirou
- Hôpital La Milétrie, Centre Hospitalier Universitaire, Poitiers, France
| | - Anthony Levy
- Hôpital La Milétrie, Centre Hospitalier Universitaire, Poitiers, France
| | - Paul Franques
- Hôpital La Milétrie, Centre Hospitalier Universitaire, Poitiers, France
| | - Hélène Gardeney
- Hôpital La Milétrie, Centre Hospitalier Universitaire, Poitiers, France
| | | | | | - Monia Ouali
- Institut de Recherche Pierre Fabre, Toulouse, France
| | | | - Pierre Ferre
- Institut de Recherche Pierre Fabre, Toulouse, France
| | | | - Michel Attal
- Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France
| | - Thierry Facon
- Service des Maladies du Sang, Centre Hospitalier Régional Universitaire, Lille, France
| | - Xavier Leleu
- Hôpital La Milétrie, Centre Hospitalier Universitaire, Poitiers, France.,Inserm Centre d'Investigation Clinique U1402, Centre Hospitalier Universitaire, Poitiers, France
| |
Collapse
|
24
|
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: 17] [Impact Index Per Article: 2.4] [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
|
25
|
Wieser V, Adolph TE, Enrich B, Kuliopulos A, Kaser A, Tilg H, Kaneider NC. Reversal of murine alcoholic steatohepatitis by pepducin-based functional blockade of interleukin-8 receptors. Gut 2017; 66:930-938. [PMID: 26858343 PMCID: PMC5531226 DOI: 10.1136/gutjnl-2015-310344] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 11/30/2015] [Accepted: 12/07/2015] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Alcoholic steatohepatitis is a life-threatening condition with short-term mortality up to 40%. It features hepatic neutrophil infiltration and blood neutrophilia, and may evolve from ethanol-induced breakdown of the enteric barrier and consequent bacteraemia. Signalling through CXCR1/2 G-protein-coupled-receptors (GPCRs), the interleukin (IL)-8 receptors, is critical for the recruitment and activation of neutrophils. We have developed short lipopeptides (pepducins), which inhibit post-ligand GPCR activation precisely targeting individual GPCRs. DESIGN Experimental alcoholic liver disease was induced by administering alcohol and a Lieber-DeCarli high-fat diet. CXCR1/2 GPCRs were blocked via pepducins either from onset of the experiment or after disease was fully established. Hepatic inflammatory infiltration, hepatocyte lipid accumulation and overall survival were assessed as primary outcome parameters. Neutrophil activation was assessed by myeloperoxidase activity and liver cell damage by aspartate aminotransferase and alanine aminotransferase plasma levels. Chemotaxis assays were performed to identify chemoattractant signals derived from alcohol-exposed hepatocytes. RESULTS Here, we show that experimental alcoholic liver disease is driven by CXCR1/2-dependent activation of neutrophils. CXCR1/2-specific pepducins not only protected mice from liver inflammation, weight loss and mortality associated with experimental alcoholic liver disease, but therapeutic administration cured disease and prevented further mortality in fully established disease. Hepatic neutrophil infiltration and triglyceride accumulation was abrogated by CXCR1/2 blockade. Moreover, CXCL-1 plasma levels were decreased with the pepducin therapy as was the transcription of hepatic IL-1β mRNA. CONCLUSIONS We propose that high circulating IL-8 in human alcoholic hepatitis may cause pathogenic overzealous neutrophil activation, and therapeutic blockade via pepducins merits clinical study.
Collapse
Affiliation(s)
- Verena Wieser
- Department of Internal Medicine I, Gastroenterology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria,Christian Doppler Research Laboratory for Gut Inflammation, Medical University Innsbruck, Innsbruck, Austria
| | - Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria,Division of Gastroenterology and Hepatology, Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Barbara Enrich
- Department of Internal Medicine I, Gastroenterology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Athan Kuliopulos
- Center for Hemostasis and Thrombosis Research, Molecular Oncology Research Institute, Tufts Medical Center, Tufts University School of Medicine, Massachusetts, USA
| | - Arthur Kaser
- Division of Gastroenterology and Hepatology, Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria,Christian Doppler Research Laboratory for Gut Inflammation, Medical University Innsbruck, Innsbruck, Austria
| | - Nicole C Kaneider
- Christian Doppler Research Laboratory for Gut Inflammation, Medical University Innsbruck, Innsbruck, Austria,Division of Gastroenterology and Hepatology, Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| |
Collapse
|
26
|
Norozi F, Shahrabi S, Hajizamani S, Saki N. Regulatory role of Megakaryocytes on Hematopoietic Stem Cells Quiescence by CXCL4/PF4 in Bone Marrow Niche. Leuk Res 2016; 48:107-12. [DOI: 10.1016/j.leukres.2015.12.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 12/26/2015] [Accepted: 12/28/2015] [Indexed: 01/20/2023]
|
27
|
Reinholdt L, Laursen MB, Schmitz A, Bødker JS, Jakobsen LH, Bøgsted M, Johnsen HE, Dybkær K. The CXCR4 antagonist plerixafor enhances the effect of rituximab in diffuse large B-cell lymphoma cell lines. Biomark Res 2016; 4:12. [PMID: 27307990 PMCID: PMC4908729 DOI: 10.1186/s40364-016-0067-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 06/08/2016] [Indexed: 12/25/2022] Open
Abstract
Background Diffuse large B-cell lymphoma (DLBCL) is an aggressive disease with variable clinical outcome, accounting for at least 25-30 % of adult non-Hodgkin lymphomas. Approximately one third of DLBCL patients are not cured by the currently used treatment regimen, R-CHOP. Hence, new treatment strategies are needed. Antagonizing the CXCR4 receptor might be promising since the CXCR4-CXCL12 axis is implicated in several aspects of tumor pathogenesis as well as in protection from chemotherapeutic response. In Burkitt lymphoma, the CXCR4 antagonist plerixafor has already been shown to enhance the therapeutic effect of rituximab, the immunotherapeutic agent of R-CHOP; but this is yet to be confirmed for DLBCL. We, therefore, investigated the effect of plerixafor on DLBCL cellular response to rituximab. Methods In this in vitro study, human DLBCL cell lines were treated with rituximab and/or plerixafor, concomitantly or in sequence. The trypan blue exclusion method and MTS-based assays were used to evaluate cellular proliferation, whereas flow cytometry was used for assessment of apoptosis status and CXCR4 surface expression level. Linear mixed effects models were used to assess statistical significance. Results We observed that simultaneous addition of plerixafor and rituximab resulted in a significant decrease in DLBCL cellular proliferation, compared to monotherapeutic response. The effect was dose-dependent, and concomitant administration was observed to be superior to sequential drug administration. Accordingly, the fraction of apoptotic/dead cells significantly increased following addition of plerixafor to rituximab treatment. Furthermore, exposure of DLBCL cells to plerixafor resulted in a significant decrease in CXCR4 fluorescence intensity. Conclusions Based on our results, implying that the anti-proliferative/pro-apoptotic effect of rituximab on DLBCL cells can be synergistically enhanced by the CXCR4 antagonist plerixafor, addition of plerixafor to the R-CHOP regimen can be suggested to improve treatment outcome for DLBCL patients. Electronic supplementary material The online version of this article (doi:10.1186/s40364-016-0067-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Linn Reinholdt
- Department of Haematology, Aalborg University Hospital, Sdr Skovvej 15, Aalborg, DK-9000 Denmark
| | - Maria Bach Laursen
- Department of Haematology, Aalborg University Hospital, Sdr Skovvej 15, Aalborg, DK-9000 Denmark
| | - Alexander Schmitz
- Department of Haematology, Aalborg University Hospital, Sdr Skovvej 15, Aalborg, DK-9000 Denmark ; Clinical Cancer Research Center, Aalborg University, Sdr Skovvej 15, Aalborg, DK-9000 Denmark
| | - Julie Støve Bødker
- Department of Haematology, Aalborg University Hospital, Sdr Skovvej 15, Aalborg, DK-9000 Denmark ; Clinical Cancer Research Center, Aalborg University, Sdr Skovvej 15, Aalborg, DK-9000 Denmark
| | - Lasse Hjort Jakobsen
- Department of Haematology, Aalborg University Hospital, Sdr Skovvej 15, Aalborg, DK-9000 Denmark ; Department of Clinical Medicine, Aalborg University, Sdr Skovvej 15, Aalborg, DK-9000 Denmark
| | - Martin Bøgsted
- Department of Haematology, Aalborg University Hospital, Sdr Skovvej 15, Aalborg, DK-9000 Denmark ; Clinical Cancer Research Center, Aalborg University, Sdr Skovvej 15, Aalborg, DK-9000 Denmark ; Department of Clinical Medicine, Aalborg University, Sdr Skovvej 15, Aalborg, DK-9000 Denmark
| | - Hans Erik Johnsen
- Department of Haematology, Aalborg University Hospital, Sdr Skovvej 15, Aalborg, DK-9000 Denmark ; Clinical Cancer Research Center, Aalborg University, Sdr Skovvej 15, Aalborg, DK-9000 Denmark ; Department of Clinical Medicine, Aalborg University, Sdr Skovvej 15, Aalborg, DK-9000 Denmark
| | - Karen Dybkær
- Department of Haematology, Aalborg University Hospital, Sdr Skovvej 15, Aalborg, DK-9000 Denmark ; Clinical Cancer Research Center, Aalborg University, Sdr Skovvej 15, Aalborg, DK-9000 Denmark ; Department of Clinical Medicine, Aalborg University, Sdr Skovvej 15, Aalborg, DK-9000 Denmark
| |
Collapse
|
28
|
Chang CH, Hale SJ, Cox CV, Blair A, Kronsteiner B, Grabowska R, Zhang Y, Cook D, Khoo CP, Schrader JB, Kabuga SB, Martin-Rendon E, Watt SM. Junctional Adhesion Molecule-A Is Highly Expressed on Human Hematopoietic Repopulating Cells and Associates with the Key Hematopoietic Chemokine Receptor CXCR4. Stem Cells 2016; 34:1664-78. [PMID: 26866290 DOI: 10.1002/stem.2340] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 01/11/2016] [Indexed: 12/16/2022]
Abstract
Hematopoietic stem/progenitor cells (HSPCs) reside in specialized bone marrow microenvironmental niches, with vascular elements (endothelial/mesenchymal stromal cells) and CXCR4-CXCL12 interactions playing particularly important roles for HSPC entry, retention, and maintenance. The functional effects of CXCL12 are dependent on its local concentration and rely on complex HSPC-niche interactions. Two Junctional Adhesion Molecule family proteins, Junctional Adhesion Molecule-B (JAM)-B and JAM-C, are reported to mediate HSPC-stromal cell interactions, which in turn regulate CXCL12 production by mesenchymal stromal cells (MSCs). Here, we demonstrate that another JAM family member, JAM-A, is most highly expressed on human hematopoietic stem cells with in vivo repopulating activity (p < .01 for JAM-A(high) compared to JAM-A(Int or Low) cord blood CD34(+) cells). JAM-A blockade, silencing, and overexpression show that JAM-A contributes significantly (p < .05) to the adhesion of human HSPCs to IL-1β activated human bone marrow sinusoidal endothelium. Further studies highlight a novel association of JAM-A with CXCR4, with these molecules moving to the leading edge of the cell upon presentation with CXCL12 (p < .05 compared to no CXCL12). Therefore, we hypothesize that JAM family members differentially regulate CXCR4 function and CXCL12 secretion in the bone marrow niche. Stem Cells 2016;34:1664-1678.
Collapse
Affiliation(s)
- Chao-Hui Chang
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | - Sarah J Hale
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | - Charlotte V Cox
- Bristol Institute for Transfusion Sciences, NHS Blood and Transplant, Bristol, United Kingdom.,Cancer Research School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Allison Blair
- Bristol Institute for Transfusion Sciences, NHS Blood and Transplant, Bristol, United Kingdom.,Cancer Research School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Barbara Kronsteiner
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | - Rita Grabowska
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | - Youyi Zhang
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | - David Cook
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | - Cheen P Khoo
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | - Jack B Schrader
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | - Suranahi Buglass Kabuga
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | - Enca Martin-Rendon
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | - Suzanne M Watt
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| |
Collapse
|
29
|
Peng SB, Zhang X, Paul D, Kays LM, Ye M, Vaillancourt P, Dowless M, Stancato LF, Stewart J, Uhlik MT, Long H, Chu S, Obungu VH. Inhibition of CXCR4 by LY2624587, a Fully Humanized Anti-CXCR4 Antibody Induces Apoptosis of Hematologic Malignancies. PLoS One 2016; 11:e0150585. [PMID: 26954567 PMCID: PMC4782998 DOI: 10.1371/journal.pone.0150585] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 02/16/2016] [Indexed: 12/31/2022] Open
Abstract
SDF-1 and CXCR4 are a chemokine and chemokine receptor pair playing critical roles in tumorigenesis. Overexpression of 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. In this study, we developed a humanized anti-CXCR4 monoclonal antibody, LY2624587 as a potent CXCR4 antagonist that was advanced into clinical study for cancer. LY2624587 blocked SDF-1 binding to CXCR4 with an IC50 of 0.26 nM, and inhibited SDF-1-induced GTP binding with a Kb of 0.66 nM. In human lymphoma U937 and leukemia CCRF-CEM cells expressing endogenous CXCR4, LY2624587 inhibited SDF-1-induced cell migration with IC50 values of 3.7 and 0.26 nM, respectively. This antibody also inhibited CXCR4 and SDF-1 mediated cell signaling including activation of MAPK and AKT in tumor cells expressing CXCR4. Bifocal microscopic and flow cytometry analyses revealed that LY2624587 mediated receptor internalization and caused CXCR4 down-regulation on the cell surface. In human hematologic cancer cells, LY2624587 caused dose dependent apoptosis in vitro and in vivo. In mouse xenograft models developed with human leukemia and lymphoma cells expressing high levels of CXCR4, LY2624587 exhibited dose-dependent tumor growth inhibition and provided significant survival benefit in a disseminated lymphoma model. Collectively, we have demonstrated that CXCR4 inhibition by LY2624587 has the potential for the treatment of human hematological malignancies.
Collapse
Affiliation(s)
- Sheng-Bin Peng
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana, 46285, United States of America
| | - Xiaoyi Zhang
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana, 46285, United States of America
| | - Donald Paul
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana, 46285, United States of America
| | - Lisa M Kays
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana, 46285, United States of America
| | - Ming Ye
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana, 46285, United States of America
| | - Peter Vaillancourt
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana, 46285, United States of America
| | - Michele Dowless
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana, 46285, United States of America
| | - Louis F Stancato
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana, 46285, United States of America
| | - Julie Stewart
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana, 46285, United States of America
| | - Mark T Uhlik
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana, 46285, United States of America
| | - Haiyan Long
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana, 46285, United States of America
| | - Shaoyou Chu
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana, 46285, United States of America
| | - Victor H Obungu
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana, 46285, United States of America
| |
Collapse
|
30
|
Regulation of the Melanocortin-Sensitive Adenylate Cyclase System by N-Acylated Peptide 71-82 of Type 4 Melanocortin Receptor. Bull Exp Biol Med 2015; 160:40-4. [PMID: 26593415 DOI: 10.1007/s10517-015-3093-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Indexed: 10/22/2022]
Abstract
The peptides structurally corresponding in to cytoplasmic loops of G protein-coupled receptors (GPCR) are able to control functional activity of homologous receptors and the corresponding signaling pathways. Modification of these peptides with hydrophobic radicals enhances their biological activity due to penetration of lipophilic derivatives through the membrane and anchoring near their targets, GPCR. We synthesized an N-palmitoylated peptide Palm-Val-[Lys-Asn-Lys-Asn-Leu-His-Ser-Pro-(Nle)-Tyr-Phe-Phe71-82]-amide-Palm-Val-(71-82) structurally corresponding to cytoplasmic loop 1 of melanocortin 4 receptor (M4R). We found that in micromolar concentrations it very effectively suppresses stimulation of basal adenylate cyclase activity and basal level of GppNHp binding of heterotrimeric G proteins produced by THIQ and α-melanocyte stimulating hormone (α-MSH), agonists of M4R homologous to the peptide, in synaptosomal membranes of rat brain. The peptide Palm-Val-(71-82) also reduced, albeit to a significantly less extent, stimulation of adenylate cyclase and G-proteins by M3R agonist of γ-MSH, due to high homology of the peptide primary structure to M3R cytoplasmic loop 1. The synthesized peptide with activity of M4R/M3R antagonist can be used for the development of regulators of M4R and M3R and the corresponding biochemical and physiological processes.
Collapse
|
31
|
Jhaveri A, Torchilin V. Intracellular delivery of nanocarriers and targeting to subcellular organelles. Expert Opin Drug Deliv 2015; 13:49-70. [DOI: 10.1517/17425247.2015.1086745] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
32
|
Zhang P, Leger AJ, Baleja JD, Rana R, Corlin T, Nguyen N, Koukos G, Bohm A, Covic L, Kuliopulos A. Allosteric Activation of a G Protein-coupled Receptor with Cell-penetrating Receptor Mimetics. J Biol Chem 2015; 290:15785-15798. [PMID: 25934391 DOI: 10.1074/jbc.m115.636316] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Indexed: 01/09/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are remarkably versatile signaling systems that are activated by a large number of different agonists on the outside of the cell. However, the inside surface of the receptors that couple to G proteins has not yet been effectively modulated for activity or treatment of diseases. Pepducins are cell-penetrating lipopeptides that have enabled chemical and physical access to the intracellular face of GPCRs. The structure of a third intracellular (i3) loop agonist, pepducin, based on protease-activated receptor-1 (PAR1) was solved by NMR and found to closely resemble the i3 loop structure predicted for the intact receptor in the on-state. Mechanistic studies revealed that the pepducin directly interacts with the intracellular H8 helix region of PAR1 and allosterically activates the receptor through the adjacent (D/N)PXXYYY motif through a dimer-like mechanism. The i3 pepducin enhances PAR1/Gα subunit interactions and induces a conformational change in fluorescently labeled PAR1 in a very similar manner to that induced by thrombin. As pepducins can potentially be made to target any GPCR, these data provide insight into the identification of allosteric modulators to this major drug target class.
Collapse
Affiliation(s)
- Ping Zhang
- From the Center of Hemostasis and Thrombosis Research, Molecular Oncology Research Institute, Tufts Medical Center, and Departments of Biochemistry and Medicine, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Andrew J Leger
- From the Center of Hemostasis and Thrombosis Research, Molecular Oncology Research Institute, Tufts Medical Center, and Departments of Biochemistry and Medicine, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - James D Baleja
- From the Center of Hemostasis and Thrombosis Research, Molecular Oncology Research Institute, Tufts Medical Center, and Departments of Biochemistry and Medicine, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Rajashree Rana
- From the Center of Hemostasis and Thrombosis Research, Molecular Oncology Research Institute, Tufts Medical Center, and Departments of Biochemistry and Medicine, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Tiffany Corlin
- From the Center of Hemostasis and Thrombosis Research, Molecular Oncology Research Institute, Tufts Medical Center, and Departments of Biochemistry and Medicine, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Nga Nguyen
- From the Center of Hemostasis and Thrombosis Research, Molecular Oncology Research Institute, Tufts Medical Center, and Departments of Biochemistry and Medicine, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Georgios Koukos
- From the Center of Hemostasis and Thrombosis Research, Molecular Oncology Research Institute, Tufts Medical Center, and Departments of Biochemistry and Medicine, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Andrew Bohm
- From the Center of Hemostasis and Thrombosis Research, Molecular Oncology Research Institute, Tufts Medical Center, and Departments of Biochemistry and Medicine, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Lidija Covic
- From the Center of Hemostasis and Thrombosis Research, Molecular Oncology Research Institute, Tufts Medical Center, and Departments of Biochemistry and Medicine, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Athan Kuliopulos
- From the Center of Hemostasis and Thrombosis Research, Molecular Oncology Research Institute, Tufts Medical Center, and Departments of Biochemistry and Medicine, Tufts University School of Medicine, Boston, Massachusetts 02111.
| |
Collapse
|
33
|
Shpakov AO, Shpakova EA. [Prospects for use of peptides and their derivatives, structurally corresponding to the G protein-coupled receptors, in medicine]. BIOCHEMISTRY MOSCOW-SUPPLEMENT SERIES B-BIOMEDICAL CHEMISTRY 2015; 61:19-29. [PMID: 25762596 DOI: 10.1134/s1990750814010144] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The regulation of signaling pathways involved in the control of many physiological functions is carried out via the heterotrimeric G protein-coupled receptors (GPCR). The search of effective and selective regulators of GPCR and intracellular signaling cascades coupled with them is one of the important problems of modern fundamental and clinical medicine. Recently data suggest that synthetic peptides and their derivatives, structurally corresponding to the intracellular and transmembrane regions of GPCR, can interact with high efficiency and selectivity with homologous receptors and influence, thus, the functional activity of intracellular signaling cascades and fundamental cellular processes controlled by them. GPCR-peptides are active in both in vitro and in vivo. They regulate hematopoiesis, angiogenesis and cell proliferation, inhibit tumor growth and metastasis, and prevent the inflammatory diseases and septic shock. These data show greatest prospects in the development of the new generations of drugs based on GPCR-derived peptides, capable of regulating the important functions of the organism.
Collapse
|
34
|
The tumor microenvironment shapes hallmarks of mature B-cell malignancies. Oncogene 2015; 34:4673-82. [PMID: 25639873 DOI: 10.1038/onc.2014.403] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 10/30/2014] [Indexed: 02/06/2023]
Abstract
B-cell tumorigenesis results from a host of known and unknown genetic anomalies, including non-random translocations of genes that normally function as determinants of cell proliferation or cell survival to regions juxtaposed to active immunoglobulin heavy chain enhancer elements, chromosomal aneuploidy, somatic mutations that further affect oncogenic signaling and loss of heterozygosity of tumor-suppressor genes. However, it is critical to recognize that even in the setting of a genetic disease, the B-cell/plasma cell tumor microenvironment (TME) contributes significantly to malignant transformation and pathogenesis. Over a decade ago, we proposed the concept of cell adhesion-mediated drug resistance to delineate a form of TME-mediated drug resistance that protects hematopoietic tumor cells from the initial effect of diverse therapies. In the interim, it has been increasingly appreciated that TME also contributes to tumor initiation and progression through sustained growth/proliferation, self-renewal capacity, immune evasion, migration and invasion as well as resistance to cell death in a host of B-cell malignancies, including mantle cell lymphoma, diffuse large B-cell lymphoma, Waldenstroms macroglobulinemia, chronic lymphocytic leukemia and multiple myeloma. Within this review, we propose that TME and the tumor co-evolve as a consequence of bidirectional signaling networks. As such, TME represents an important target and should be considered integral to tumor progression and drug response.
Collapse
|
35
|
Abstract
Lipopeptides based on the intracellular loops of cell-surface receptors, known as "Pepducins," represent a promising new class of compounds used for the study of membrane proteins and as potential therapeutics in a variety of diseases. Detailed knowledge of the three-dimensional structure of G-protein-coupled receptors (GPCRs) and delineation of the mechanisms of pepducin activation and biased G-protein signaling has facilitated the development of even more potent pepducin allosteric modulators.
Collapse
|
36
|
Moreno MJ, Bosch R, Dieguez-Gonzalez R, Novelli S, Mozos A, Gallardo A, Pavón MÁ, Céspedes MV, Grañena A, Alcoceba M, Blanco O, Gonzalez-Díaz M, Sierra J, Mangues R, Casanova I. CXCR4 expression enhances diffuse large B cell lymphoma dissemination and decreases patient survival. J Pathol 2014; 235:445-55. [PMID: 25231113 DOI: 10.1002/path.4446] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 09/04/2014] [Accepted: 09/11/2014] [Indexed: 12/17/2022]
Abstract
The chemokine receptor CXCR4 has been implicated in the migration and trafficking of malignant B cells in several haematological malignancies. Over-expression of CXCR4 has been identified in haematological tumours, but data concerning the role of this receptor in diffuse large B cell lymphoma (DLBCL) are lacking. CXCR4 is a marker of poor prognosis in various neoplasms, correlating with metastatic disease and decreased survival of patients. We studied CXCR4 involvement in cell migration in vitro and dissemination in vivo. We also evaluated the prognostic significance of CXCR4 in 94 biopsies of DLBCL patients. We observed that the level of expression of CXCR4 in DLBCL cell lines correlated positively with in vitro migration. Expression of the receptor was also associated with increased engraftment and dissemination, and decreased survival time in NOD/SCID mice. Furthermore, administration of a specific CXCR4 antagonist, AMD3100, decreased dissemination of DLBCL cells in a xenograft mouse model. In addition, we found that CXCR4 expression is an independent prognostic factor for shorter overall survival and progression-free survival in DLBCL patients. These results show that CXCR4 mediates dissemination of DLBCL cells and define for the first time its value as an independent prognostic marker in DLBCL patients.
Collapse
Affiliation(s)
- María José Moreno
- Grup d'Oncogènesi i Antitumorals, lnstitut d'Investigacions Biomèdiques Sant Pau, Barcelona, Spain; Department of Biochemistry and Molecular Biology, Universitat de Barcelona, Spain; CIBER en Bioingeniería, Biomateriales y Nanomecidicina (CIBER-BBN), Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Reissmann S. Cell penetration: scope and limitations by the application of cell-penetrating peptides. J Pept Sci 2014; 20:760-84. [DOI: 10.1002/psc.2672] [Citation(s) in RCA: 176] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 06/06/2014] [Accepted: 06/10/2014] [Indexed: 12/23/2022]
Affiliation(s)
- Siegmund Reissmann
- Friedrich Schiller University, Biological and Pharmaceutical Faculty; Institute of Biochemistry and Biophysics; Dornburger Strasse 25 07743 Jena Germany
- Jena Bioscience GmbH; Loebstedter Strasse 80 07749 Jena Germany
| |
Collapse
|
38
|
Abstract
B cell lymphomas are cancers that arise from cells that depend on numerous highly orchestrated interactions with immune and stromal cells in the course of normal development. Despite the recent focus on dissecting the genetic aberrations within cancer cells, it has been increasingly recognized that tumour cells retain a range of dependence on interactions with the non-malignant cells and stromal elements that constitute the tumour microenvironment. A fundamental understanding of these interactions gives insight into the pathogenesis of most B cell lymphomas and, moreover, identifies novel therapeutic opportunities for targeting oncogenic pathways, both now and in the future.
Collapse
Affiliation(s)
- David W Scott
- Centre for Lymphoid Cancer, British Columbia Cancer Agency, Vancouver V5Z 1L3, Canada
| | - Randy D Gascoyne
- 1] Centre for Lymphoid Cancer, British Columbia Cancer Agency, Vancouver V5Z 1L3, Canada. [2] Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver V6T 2B5, Canada
| |
Collapse
|
39
|
Park TS, Donnenberg VS, Donnenberg AD, Zambidis ET, Zimmerlin L. Dynamic Interactions Between Cancer Stem Cells And Their Stromal Partners. CURRENT PATHOBIOLOGY REPORTS 2014; 2:41-52. [PMID: 24660130 PMCID: PMC3956651 DOI: 10.1007/s40139-013-0036-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The cancer stem cell (CSC) paradigm presumes the existence of self-renewing cancer cells capable of regenerating all tumor compartments and exhibiting stem cell-associated phenotypes. Recent interpretations of the CSC hypothesis envision stemness as a dynamic trait of tumor-initiating cells rather than a defined and unique cell type. Bidirectional crosstalk between the tumor microenvironment and the cancer bulk is well described in the literature and the tumor-associated stroma, vasculature and immune infiltrate have all been implicated as direct contributors to tumor development. These non-neoplastic cell types have also been shown to organize specific niches within the tumor bulk where they can control the intra-tumor CSC content and alter the fate of CSCs and tumor progenitors during tumorigenesis to acquire phenotypic features for invasion, metastasis and dormancy. Despite the complexity of the tumor-stroma interactome, novel therapeutic approaches envision combining tumor-ablative treatment with manipulation of the tumor microenvironment. We will review the currently available literature that provides clues about the complex cellular network that regulate the CSC phenotype and its niches during tumor progression.
Collapse
Affiliation(s)
- Tea Soon Park
- Institute for Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, United States of America
| | - Vera S. Donnenberg
- University of Pittsburgh School of Medicine, Department of Cardiothoracic Surgery, Pittsburgh, Pennsylvania, United States of America
- University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
- McGowan Institute of Regenerative Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Albert D. Donnenberg
- University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
- McGowan Institute of Regenerative Medicine, Pittsburgh, Pennsylvania, United States of America
- University of Pittsburgh School of Medicine, Department of Medicine, Division of Hematology/Oncology, Pittsburgh, Pennsylvania, United States of America
| | - Elias T. Zambidis
- Institute for Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, United States of America
| | - Ludovic Zimmerlin
- Institute for Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, United States of America
| |
Collapse
|
40
|
Pepducin targeting the C-X-C chemokine receptor type 4 acts as a biased agonist favoring activation of the inhibitory G protein. Proc Natl Acad Sci U S A 2013; 110:E5088-97. [PMID: 24309376 DOI: 10.1073/pnas.1312515110] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Short lipidated peptide sequences derived from various intracellular loop regions of G protein-coupled receptors (GPCRs) are named pepducins and act as allosteric modulators of a number of GPCRs. Recently, a pepducin selectively targeting the C-X-C chemokine receptor type 4 (CXCR4) was found to be an allosteric agonist, active in both cell-based assays and in vivo. However, the precise mechanism of action of this class of ligands remains poorly understood. In particular, given the diversity of signaling effectors that can be engaged by a given receptor, it is not clear whether pepducins can show biased signaling leading to functional selectivity. To explore the ligand-biased potential of pepducins, we assessed the effect of the CXCR4 selective pepducin, ATI-2341, on the ability of the receptor to engage the inhibitory G proteins (Gi1, Gi2 and Gi3), G13, and β-arrestins. Using bioluminescence resonance energy transfer-based biosensors, we found that, in contrast to the natural CXCR4 ligand, stromal cell-derived factor-1α, which promotes the engagement of the three Gi subtypes, G13 and the two β-arrestins, ATI-2341 leads to the engagement of the Gi subtypes but not G13 or the β-arrestins. Calculation of the transduction ratio for each pathway revealed a strong negative bias of ATI-2341 toward G13 and β-arrestins, revealing functional selectivity for the Gi pathways. The negative bias toward β-arrestins results from the reduced ability of the pepducin to promote GPCR kinase-mediated phosphorylation of the receptor. In addition to revealing ligand-biased signaling of pepducins, these findings shed some light on the mechanism of action of a unique class of allosteric regulators.
Collapse
|
41
|
Somovilla-Crespo B, Alfonso-Pérez M, Cuesta-Mateos C, Carballo-de Dios C, Beltrán AE, Terrón F, Pérez-Villar JJ, Gamallo-Amat C, Pérez-Chacón G, Fernández-Ruiz E, Zapata JM, Muñoz-Calleja C. Anti-CCR7 therapy exerts a potent anti-tumor activity in a xenograft model of human mantle cell lymphoma. J Hematol Oncol 2013; 6:89. [PMID: 24305507 PMCID: PMC3879031 DOI: 10.1186/1756-8722-6-89] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 11/19/2013] [Indexed: 12/19/2022] Open
Abstract
Background The chemokine receptor CCR7 mediates lymphoid dissemination of many cancers, including lymphomas and epithelial carcinomas, thus representing an attractive therapeutic target. Previous results have highlighted the potential of the anti-CCR7 monoclonal antibodies to inhibit migration in transwell assays. The present study aimed to evaluate the in vivo therapeutic efficacy of an anti-CCR7 antibody in a xenografted human mantle cell lymphoma model. Methods NOD/SCID mice were either subcutaneously or intravenously inoculated with Granta-519 cells, a human cell line derived from a leukemic mantle cell lymphoma. The anti-CCR7 mAb treatment (3 × 200 μg) was started on day 2 or 7 to target lymphoma cells in either a peri-implantation or a post-implantation stage, respectively. Results The anti-CCR7 therapy significantly delayed the tumor appearance and also reduced the volumes of tumors in the subcutaneous model. Moreover, an increased number of apoptotic tumor cells was detected in mice treated with the anti-CCR7 mAb compared to the untreated animals. In addition, significantly reduced number of Granta-519 cells migrated from subcutaneous tumors to distant lymphoid organs, such as bone marrow and spleen in the anti-CCR7 treated mice. In the intravenous models, the anti-CCR7 mAb drastically increased survival of the mice. Accordingly, dissemination and infiltration of tumor cells in lymphoid and non-lymphoid organs, including lungs and central nervous system, was almost abrogated. Conclusions The anti-CCR7 mAb exerts a potent anti-tumor activity and might represent an interesting therapeutic alternative to conventional therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Cecilia Muñoz-Calleja
- Instituto de Investigación Sanitaria Princesa, Department of Immunology, Hospital Universitario de La Princesa, C/Diego de León 62, Madrid 28006, Spain.
| |
Collapse
|
42
|
Amé-Thomas P, Tarte K. The yin and the yang of follicular lymphoma cell niches: role of microenvironment heterogeneity and plasticity. Semin Cancer Biol 2013; 24:23-32. [PMID: 23978491 DOI: 10.1016/j.semcancer.2013.08.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 07/31/2013] [Accepted: 08/15/2013] [Indexed: 12/20/2022]
Abstract
Follicular lymphoma (FL) results from the malignant transformation of germinal center B cells and is characterized by recurrent genetic alterations providing a direct growth advantage or facilitating interaction with tumor microenvironment. In agreement, accumulating evidences suggest a dynamic bidirectional crosstalk between FL B cells and surrounding non-malignant cells within specialized tumor niches in both invaded lymph nodes and bone marrow. Infiltrating stromal cells, macrophages, and T/NK cell subsets either contribute to anti-tumor immune response, or conversely form a tumor supportive network promoting FL B cell survival, growth, and drug resistance. This review depicts the phenotypic heterogeneity and functional plasticity of the most important FL cell partners and describes their complex interplay. We also unravel how malignant B cells recruit and subvert accessory immune and stromal cells to trigger their polarization toward a supportive phenotype. Based on these observations, innovative therapeutic approaches have been recently proposed, in order to benefit from local anti-tumor immunity and/or to selectively target the protective cell niche.
Collapse
Affiliation(s)
- Patricia Amé-Thomas
- INSERM, UMR U917, Equipe Labellisée Ligue Contre le Cancer, Faculté de Médecine, Rennes, France; Université Rennes 1, Rennes, France; CHU de Rennes, Hôpital Pontchaillou, Service ITeCH, Pôle de Biologie, Rennes, France
| | - Karin Tarte
- INSERM, UMR U917, Equipe Labellisée Ligue Contre le Cancer, Faculté de Médecine, Rennes, France; Université Rennes 1, Rennes, France; CHU de Rennes, Hôpital Pontchaillou, Service ITeCH, Pôle de Biologie, Rennes, France; Etablissement Français du Sang Bretagne, Rennes, France.
| |
Collapse
|
43
|
Beider K, Ribakovsky E, Abraham M, Wald H, Weiss L, Rosenberg E, Galun E, Avigdor A, Eizenberg O, Peled A, Nagler A. Targeting the CD20 and CXCR4 pathways in non-hodgkin lymphoma with rituximab and high-affinity CXCR4 antagonist BKT140. Clin Cancer Res 2013; 19:3495-507. [PMID: 23637121 DOI: 10.1158/1078-0432.ccr-12-3015] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Chemokine axis CXCR4/CXCL12 is critically involved in the survival and trafficking of normal and malignant B lymphocytes. Here, we investigated the effect of high-affinity CXCR4 antagonist BKT140 on lymphoma cell growth and rituximab-induced cytotoxicity in vitro and in vivo. EXPERIMENTAL DESIGN In vitro efficacy of BKT140 alone or in combination with rituximab was determined in non-Hodgkin lymphoma (NHL) cell lines and primary samples from bone marrow aspirates of patients with NHL. In vivo efficacy was evaluated in xenograft models of localized and disseminated NHL with bone marrow involvement. RESULTS Antagonizing CXCR4 with BKT140 resulted in significant inhibition of CD20+ lymphoma cell growth and in the induction of cell death, respectively. Combination of BKT140 with rituximab significantly enhanced the apoptosis against the lymphoma cells in a dose-dependent manner. Moreover, rituximab induced CXCR4 expression in lymphoma cell lines and primary lymphoma cells, suggesting the possible interaction between CD20 and CXCR4 pathways in NHL. Primary bone marrow stromal cells (BMSC) further increased CXCR4 expression and protected NHL cells from rituximab-induced apoptosis, whereas BKT140 abrogated this protective effect. Furthermore, BKT140 showed efficient antilymphoma activity in vivo in the xenograft model of disseminated NHL with bone marrow involvement. BKT140 treatment inhibited the local tumor progression and significantly reduced the number of NHL cells in the bone marrow. Combined treatment of BKT140 with rituximab further decreased the number of viable lymphoma cells in the bone marrow, achieving 93% reduction. CONCLUSIONS These findings suggest the possible role of CXCR4 in NHL progression and response to rituximab and provide the scientific basis for the development of novel CXCR4-targeted therapies for refractory NHL.
Collapse
Affiliation(s)
- Katia Beider
- Division of Hematology, Bone Marrow Transplantation and Cord Blood Bank, Sheba Medical Center, Tel-Hashomer, Israel
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Li L, Cole J, Margolin DA. Cancer stem cell and stromal microenvironment. Ochsner J 2013; 13:109-118. [PMID: 23531695 PMCID: PMC3603173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023] Open
Abstract
BACKGROUND Chemotherapeutic resistance and local recurrence or distant organ metastasis are the major causes of cancer mortality. Conventional cancer treatments do not consistently prevent cancer recurrence. METHODS We illustrate the key roles that cancer stem cells and the tumor microenvironment-particularly the lymph node stromal microenvironment-play in tumor drug resistance, metastasis, and recurrence in 2 representative cancers: colorectal cancer and follicular lymphoma. CONCLUSION We believe that combination treatment with chemotherapeutic agents in conjunction with targeted therapies, such as stromal/cancer stem cell signaling-targeted therapy, may effectively minimize cancer recurrence.
Collapse
Affiliation(s)
- Li Li
- Institute for Translational Research, Laboratory of Translational Cancer Research
| | - John Cole
- Department of Hematology and Oncology
- The University of Queensland School of Medicine, Ochsner Clinical School, New Orleans, LA
| | - David A. Margolin
- Department of Colon and Rectal Surgery, Ochsner Clinic Foundation, and
- The University of Queensland School of Medicine, Ochsner Clinical School, New Orleans, LA
| |
Collapse
|
45
|
Schrader A, Meyer K, von Bonin F, Vockerodt M, Walther N, Hand E, Ulrich A, Matulewicz K, Lenze D, Hummel M, Kieser A, Engelke M, Trümper L, Kube D. Global gene expression changes of in vitro stimulated human transformed germinal centre B cells as surrogate for oncogenic pathway activation in individual aggressive B cell lymphomas. Cell Commun Signal 2012; 10:43. [PMID: 23253402 PMCID: PMC3566944 DOI: 10.1186/1478-811x-10-43] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 11/25/2012] [Indexed: 12/25/2022] Open
Abstract
Background Aggressive Non-Hodgkin lymphomas (NHL) are a group of lymphomas derived from germinal centre B cells which display a heterogeneous pattern of oncogenic pathway activation. We postulate that specific immune response associated signalling, affecting gene transcription networks, may be associated with the activation of different oncogenic pathways in aggressive Non-Hodgkin lymphomas (NHL). Methodology The B cell receptor (BCR), CD40, B-cell activating factor (BAFF)-receptors and Interleukin (IL) 21 receptor and Toll like receptor 4 (TLR4) were stimulated in human transformed germinal centre B cells by treatment with anti IgM F(ab)2-fragments, CD40L, BAFF, IL21 and LPS respectively. The changes in gene expression following the activation of Jak/STAT, NF-кB, MAPK, Ca2+ and PI3K signalling triggered by these stimuli was assessed using microarray analysis. The expression of top 100 genes which had a change in gene expression following stimulation was investigated in gene expression profiles of patients with Aggressive non-Hodgkin Lymphoma (NHL). Results αIgM stimulation led to the largest number of changes in gene expression, affecting overall 6596 genes. While CD40L stimulation changed the expression of 1194 genes and IL21 stimulation affected 902 genes, only 283 and 129 genes were modulated by lipopolysaccharide or BAFF receptor stimulation, respectively. Interestingly, genes associated with a Burkitt-like phenotype, such as MYC, BCL6 or LEF1, were affected by αIgM. Unique and shared gene expression was delineated. NHL-patients were sorted according to their similarity in the expression of TOP100 affected genes to stimulated transformed germinal centre B cells The αIgM gene module discriminated individual DLBCL in a similar manner to CD40L or IL21 gene modules. DLBCLs with low module activation often carry chromosomal MYC aberrations. DLBCLs with high module activation show strong expression of genes involved in cell-cell communication, immune responses or negative feedback loops. Using chemical inhibitors for selected kinases we show that mitogen activated protein kinase- and phosphoinositide 3 kinase-signalling are dominantly involved in regulating genes included in the αIgM gene module. Conclusion We provide an in vitro model system to investigate pathway activation in lymphomas. We defined the extent to which different immune response associated pathways are responsible for differences in gene expression which distinguish individual DLBCL cases. Our results support the view that tonic or constitutively active MAPK/ERK pathways are an important part of oncogenic signalling in NHL. The experimental model can now be applied to study the therapeutic potential of deregulated oncogenic pathways and to develop individual treatment strategies for lymphoma patients.
Collapse
Affiliation(s)
- Alexandra Schrader
- Department of Haematology and Oncology, University Medical Centre Göttingen, Göttingen, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Current world literature. Curr Opin Oncol 2012; 24:756-68. [PMID: 23079785 DOI: 10.1097/cco.0b013e32835a4c91] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
47
|
Mourcin F, Pangault C, Amin-Ali R, Amé-Thomas P, Tarte K. Stromal cell contribution to human follicular lymphoma pathogenesis. Front Immunol 2012; 3:280. [PMID: 22973275 PMCID: PMC3433684 DOI: 10.3389/fimmu.2012.00280] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 08/16/2012] [Indexed: 12/19/2022] Open
Abstract
Follicular lymphoma (FL) is the prototypical model of indolent B cell lymphoma displaying a strong dependence on a specialized cell microenvironment mimicking normal germinal center. Within malignant cell niches in invaded lymph nodes and bone marrow, external stimuli provided by infiltrating stromal cells make a pivotal contribution to disease development, progression, and drug resistance. The crosstalk between FL B cells and stromal cells is bidirectional, causing activation of both partners. In agreement, FL stromal cells exhibit specific phenotypic, transcriptomic, and functional properties. This review highlights the critical pathways involved in the direct tumor-promoting activity of stromal cells but also their role in the organization of FL cell niche through the recruitment of accessory immune cells and their polarization to a B cell supportive phenotype. Finally, deciphering the interplay between stromal cells and FL cells provides potential new therapeutic targets with the aim to mobilize malignant cells outside their protective microenvironment and increase their sensitivity to conventional treatment.
Collapse
|
48
|
Crews LA, Jamieson CHM. Selective elimination of leukemia stem cells: hitting a moving target. Cancer Lett 2012; 338:15-22. [PMID: 22906415 DOI: 10.1016/j.canlet.2012.08.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 07/27/2012] [Accepted: 08/07/2012] [Indexed: 01/02/2023]
Abstract
Despite the widespread use of chemotherapeutic cytotoxic agents that eradicate proliferating cell populations, patients suffering from a wide variety of malignancies continue to relapse as a consequence of resistance to standard therapies. In hematologic malignancies, leukemia stem cells (LSCs) represent a malignant reservoir of disease that is believed to drive relapse and resistance to chemotherapy and tyrosine kinase inhibitor (TKIs). Major research efforts in recent years have been aimed at identifying and characterizing the LSC population in leukemias, such as chronic myeloid leukemia (CML), which represents an important paradigm for understanding the molecular evolution of cancer. However, the precise molecular mechanisms that promote LSC-mediated therapeutic recalcitrance have remained elusive. It has become clear that the LSC population evolves during disease progression, thus presenting a serious challenge for development of effective therapeutic strategies. Multiple reports have demonstrated that LSC initiation and propagation occurs as a result of aberrant activation of pro-survival and self-renewal pathways regulated by stem-cell related signaling molecules including β-catenin and Sonic Hedgehog (Shh). Enhanced survival in LSC protective microenvironments, such as the bone marrow niche, as well as acquired dormancy of cells in these niches, also contributes to LSC persistence. Key components of these cell-intrinsic and cell-extrinsic pathways provide novel potential targets for therapies aimed at eradicating this dynamic and therapeutically recalcitrant LSC population. Furthermore, combination strategies that exploit LSC have the potential to dramatically improve the quality and quantity of life for patients that are resistant to current therapies.
Collapse
Affiliation(s)
- Leslie A Crews
- Department of Medicine, Stem Cell Program and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | | |
Collapse
|
49
|
Lee CG, Das B, Lin TL, Grimes C, Zhang X, Lavezzi T, Huang L, Cole J, Yau L, Li1 L. A rare fraction of drug-resistant follicular lymphoma cancer stem cells interacts with follicular dendritic cells to maintain tumourigenic potential. Br J Haematol 2012; 158:79-90. [PMID: 22509798 PMCID: PMC3374069 DOI: 10.1111/j.1365-2141.2012.09123.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Accepted: 03/06/2012] [Indexed: 12/25/2022]
Abstract
Follicular lymphoma (FL) comprises nearly 25% of non-Hodgkin lymphoma cases and is clinically characterized by initial sensitivity to chemotherapy followed by relapse. FL stroma contains a special type of stromal cell found in the germinal centre of lymph nodes-the follicular dendritic cell (FDC). We first isolated tumourigenic cells from the FL cell line FLK-1 by side population (SP) technique, and found that SP cells, which express ABCG2, were enriched by chemotherapy and radiation treatments. In vitro, SP cells were attracted by and adhered to FDCs through chemokine (C-X-C motif) ligand 12/chemokine (C-X-C motif) receptor 4 (CXCL12/CXCR4) signalling. In vivo, limiting dilution assays showed SP cells were highly enriched in cancer stem cells (CSC), but required FDC for tumour formation in non-obese diabetic/severe combined immunodeficiency mice. Treatment with AMD3100, a specific CXCL12/CXCR4 inhibitor, eliminated tumour growth. These findings were then verified with FL cells isolated from an FL patient's ascitic fluid (FLA-1). Finally, we detected the ABCG2 expressing lymphoma cells in FL clinical specimens. Thus, we found that the highly tumourigenic FL cells having CSC-like activities (FL-SC) interact with FDCs in a CXCL12/CXCR4 dependent manner to resist chemotherapy. Our results indicate the importance of FL-SC and niche cell signalling in maintaining tumourigenicity. These signals represent novel targets for CSC eradication.
Collapse
MESH Headings
- Animals
- Cell Communication/immunology
- Cell Line, Tumor
- Chemokine CXCL12/immunology
- Dendritic Cells, Follicular/immunology
- Dendritic Cells, Follicular/metabolism
- Dendritic Cells, Follicular/pathology
- Drug Resistance, Neoplasm
- Female
- Humans
- Immunohistochemistry
- Lymphoma, Follicular/immunology
- Lymphoma, Follicular/metabolism
- Lymphoma, Follicular/pathology
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Neoplastic Stem Cells/immunology
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Receptors, CXCR4/immunology
- Signal Transduction
- Stromal Cells/immunology
- Stromal Cells/metabolism
- Stromal Cells/pathology
Collapse
Affiliation(s)
- Chung-Gi Lee
- Laboratory of Cellular Immunology, Ochsner Clinic Foundation, New Orleans, Louisiana, USA
| | - Bikul Das
- Department of Medical Oncology, Stanford University Medical School, Stanford, California, USA
| | - Tara L. Lin
- Division of Hematology and Oncology, University of Kansas, Kansas City, Kansas, USA
| | - Chelsea Grimes
- Laboratory of Cellular Immunology, Ochsner Clinic Foundation, New Orleans, Louisiana, USA
| | - Xin Zhang
- Laboratory of Cellular Immunology, Ochsner Clinic Foundation, New Orleans, Louisiana, USA
| | - Tracey Lavezzi
- Laboratory of Cellular Immunology, Ochsner Clinic Foundation, New Orleans, Louisiana, USA
| | - Li Huang
- Department of Pathology, Ochsner Clinic Foundation, New Orleans, Louisiana, USA
| | - John Cole
- Department of Hematology and Oncology, Ochsner Clinic Foundation, New Orleans, Louisiana, USA
| | - Lillian Yau
- Department of Biostatistics, Tulane University, New Orleans, Louisiana, USA
| | - Li Li1
- Laboratory of Cellular Immunology, Ochsner Clinic Foundation, New Orleans, Louisiana, USA
| |
Collapse
|
50
|
Höpken UE, Rehm A. Homeostatic chemokines guide lymphoma cells to tumor growth-promoting niches within secondary lymphoid organs. J Mol Med (Berl) 2012; 90:1237-45. [PMID: 22577036 DOI: 10.1007/s00109-012-0906-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 04/23/2012] [Accepted: 04/26/2012] [Indexed: 02/27/2023]
Abstract
The interaction between lymphoid tumor cells and their tissue microenvironment is thought to promote dissemination and progression of lymphoma. Those type of interactions consists of at least three cornerstones, among them mesenchymal- or bone marrow-derived stromal cells, cells of the innate or adaptive immune response, and the lymphoma cells themselves. The molecular pathways of crosstalk between the lymphoma cells and their nursing stroma are not well understood and their dissection is challenging because of (1) the complexity of stroma cell subpopulations, (2) kinetic and developmental transitions/switches of stroma composition, and (3) inherent technical difficulties to isolate and analyze defined stroma cell subsets. However, recent studies of bone marrow stroma interaction with leukemia or lymphoma cells have revealed therapeutic targets involved in regulating tumor cell mobilization. Release of tumor cells from their supportive niches could be effectuated by inhibition of homing and retention signals. The present review focuses on the effects of homing receptors and cytokines attributed to lymphoid tissue formation in tumor-stroma interactions within secondary lymphoid tissues. We discuss possible cellular and molecular mechanisms of lymphoma-stroma crosstalk and highlight novel therapeutic strategies based on the disruption of tumor-stroma interaction in secondary lymphoid organs.
Collapse
Affiliation(s)
- Uta E Höpken
- Department of Tumor Genetics and Immunogenetics, Max-Delbrück-Center for Molecular Medicine, MDC, Berlin 13125, Germany.
| | | |
Collapse
|