51
|
Stepanov AV, Rybinets AS, Dronina MA, Deev SM. Study of Fibronectin Type III-Like Domains Role in Activation of gp130 Receptor. Bull Exp Biol Med 2016; 161:72-4. [PMID: 27270939 DOI: 10.1007/s10517-016-3348-8] [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: 05/03/2015] [Indexed: 11/30/2022]
Abstract
Chimeric gp130 receptors were produced to study the role of three fibronectin type III-like domains in activation of gp130 receptor machinery. The ligand-induced dimerization of gp130 was sufficient to trigger STAT3 signaling pathway. These findings can be used as the basis in designing novel therapeutic gp130 inhibitors.
Collapse
Affiliation(s)
- A V Stepanov
- M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.
| | - A S Rybinets
- M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - M A Dronina
- M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - S M Deev
- M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
52
|
GAO JIAN, ZHAO SEN, HALSTENSEN TRONDS. Increased interleukin-6 expression is associated with poor prognosis and acquired cisplatin resistance in head and neck squamous cell carcinoma. Oncol Rep 2016; 35:3265-74. [PMID: 27108527 PMCID: PMC4869939 DOI: 10.3892/or.2016.4765] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 12/31/2015] [Indexed: 12/14/2022] Open
Abstract
Increased expression of interleukin 6 (IL-6) is associated with poor prognosis and chemoresistance in many different carcinomas, but its role in head and neck squamous cell carcinoma (HNSCC) is still unsettled. Analyzing tumorous mRNA expression data from 399 HNSCC patients revealed that high IL-6 expression predicted poor prognosis. Similar tendency was observed in platinum treated patients, suggesting an IL-6 associated cisplatin resistance. IL-6 increase was also found in two in-house acquired cisplatin‑resistant HNSCC cell lines (both basaloid and conventional squamous cell carcinoma) by using microarray analysis. However, although the in-house acquired cisplatin-resistant cell lines had higher basal and markedly increased cisplatin-induced IL-6 expression, IL-6 did not mediate the cisplatin resistance as neither exogenous IL-6 nor IL-6R/gp130 inhibitors affected cisplatin sensitivity. Moreover, the IL-6/STAT3 pathway was impaired in the resistant cell lines, partly due to decreased IL-6R expression. Thus, high IL-6 expression correlated to poor prognosis and acquired cisplatin resistance, but it did not mediate cisplatin resistance in the HNSCC cell lines.
Collapse
Affiliation(s)
- JIAN GAO
- Department of Oral Biology, University of Oslo, Blindern, Oslo 0316, Norway
| | - SEN ZHAO
- Genome Biology Group, Department of Molecular Oncology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo 0424, Norway
| | | |
Collapse
|
53
|
Mastorci K, Muraro E, Pasini E, Furlan C, Sigalotti L, Cinco M, Dolcetti R, Fratta E. Toll-Like Receptor 1/2 and 5 Ligands Enhance the Expression of Cyclin D1 and D3 and Induce Proliferation in Mantle Cell Lymphoma. PLoS One 2016; 11:e0153823. [PMID: 27123851 PMCID: PMC4849792 DOI: 10.1371/journal.pone.0153823] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 04/04/2016] [Indexed: 12/28/2022] Open
Abstract
Mantle cell lymphoma (MCL) is an aggressive B-cell non-Hodgkin’s lymphoma with a still undefined etiology. Several lines of evidence are consistent with the possible involvement of peculiar microenvironmental stimuli sustaining tumor cell growth and survival, as the activation of Toll-like receptors (TLR) 4 and 9. However, little is known about the contribution of other TLRs of pathogenic relevance in the development of MCL. This study reports evidence that MCL cell lines and primary MCL cells express different levels of TLR2 and TLR5, and that their triggering is able to further activate the Akt, MAPK, and NF-κB signaling cascades, known to be altered in MCL cells. This leads to the enhancement of cyclin D1 and D3 over-expression, occurring at post-translational level through a mechanism that likely involves the Akt/GSK-3α/β pathway. Interestingly, in primary B cells, TLR1/2 or TLR5 ligands increase protein level of cyclin D1, which is not usually expressed in normal B cells, and cyclin D3 when associated with CD40 ligand (CD40L), IL-4, and anti-human-IgM co-stimulus. Finally, the activation of TLR1/2 and TLR5 results in an increased proliferation of MCL cell lines and, in the presence of co-stimulation with CD40L, IL-4, and anti-human-IgM also of primary MCL cells and normal B lymphocytes. These effects befall together with an enhanced IL-6 production in primary cultures. Overall, our findings suggest that ligands for TLR1/2 or TLR5 may provide critical stimuli able to sustain the growth and the malignant phenotype of MCL cells. Further studies aimed at identifying the natural source of these TLR ligands and their possible pathogenic association with MCL are warranted in order to better understand MCL development, but also to define new therapeutic targets for counteracting the tumor promoting effects of lymphoma microenvironment.
Collapse
Affiliation(s)
- Katy Mastorci
- Cancer Bio-Immunotherapy Unit, Department of Translational Research, Centro di Riferimento Oncologico, IRCCS—National Cancer Institute, Aviano (PN), Italy
| | - Elena Muraro
- Cancer Bio-Immunotherapy Unit, Department of Translational Research, Centro di Riferimento Oncologico, IRCCS—National Cancer Institute, Aviano (PN), Italy
- * E-mail:
| | - Elisa Pasini
- Cancer Bio-Immunotherapy Unit, Department of Translational Research, Centro di Riferimento Oncologico, IRCCS—National Cancer Institute, Aviano (PN), Italy
- Princess Margaret Cancer Centre, University Health Network and TECHNA Institute for the Advancement of Technology for Health, TMDT, Room 11–314, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Chiara Furlan
- Cancer Bio-Immunotherapy Unit, Department of Translational Research, Centro di Riferimento Oncologico, IRCCS—National Cancer Institute, Aviano (PN), Italy
| | - Luca Sigalotti
- Cancer Bio-Immunotherapy Unit, Department of Translational Research, Centro di Riferimento Oncologico, IRCCS—National Cancer Institute, Aviano (PN), Italy
| | - Marina Cinco
- Spirochete Laboratory, Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Riccardo Dolcetti
- Cancer Bio-Immunotherapy Unit, Department of Translational Research, Centro di Riferimento Oncologico, IRCCS—National Cancer Institute, Aviano (PN), Italy
- University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Australia
| | - Elisabetta Fratta
- Cancer Bio-Immunotherapy Unit, Department of Translational Research, Centro di Riferimento Oncologico, IRCCS—National Cancer Institute, Aviano (PN), Italy
- * E-mail:
| |
Collapse
|
54
|
Iqbal J, Naushad H, Bi C, Yu J, Bouska A, Rohr J, Chao W, Fu K, Chan WC, Vose JM. Genomic signatures in B-cell lymphoma: How can these improve precision in diagnosis and inform prognosis? Blood Rev 2016; 30:73-88. [DOI: 10.1016/j.blre.2015.08.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 08/07/2015] [Accepted: 08/10/2015] [Indexed: 01/07/2023]
|
55
|
Abstract
Mantle cell lymphoma (MCL) is a rare and aggressive form of non-Hodgkin lymphoma. Ibrutinib is a first-in-class, oral inhibitor of Bruton's tyrosine kinase which acts by downstream inhibition of the B-cell receptor. Early clinical trials have demonstrated excellent tolerability and a modest side-effect profile in relapsed/refractory MCL. Although the majority of disease responses are partial, efficacy data are impressive with more than two-thirds of patients demonstrating a durable response. This article focuses on all aspects of ibrutinib in the context of MCL, including a summary of the basic pharmacology and pharmacokinetics; a review of the safety and efficacy data published to date and a discussion of the future implications in MCL.
Collapse
Affiliation(s)
- David L Tucker
- Department of Haematology, Plymouth Hospitals NHS Trust, Plymouth, UK
| | - Simon A Rule
- Department of Haematology, Plymouth Hospitals NHS Trust, Plymouth, UK
| |
Collapse
|
56
|
Abstract
Mantle cell lymphoma (MCL) is an uncommon subtype of non-Hodgkin lymphoma previously considered to have a poor prognosis. Large gains were made in the first decade of the new century when clinical trials established the importance of high-dose therapy and autologous stem-cell rescue and high-dose cytarabine in younger patients and the benefits of maintenance rituximab and bendamustine in older patients. In particular, greater depth of understanding of the molecular pathophysiology of MCL has resulted in an explosion of specifically targeted new efficacious agents. In particular, agents recently approved by the Food and Drug Administration include the proteasome inhibitor bortezomib, immunomodulator lenalidomide, and Bruton's tyrosine kinase inhibitor ibrutinib. We review recent advances in the understanding of MCL biology and outline our recommended approach to therapy, including choice of chemoimmunotherapy, the role of stem-cell transplantation, and mechanism-based targeted therapies, on the basis of a synthesis of the data from published clinical trials.
Collapse
Affiliation(s)
- Chan Yoon Cheah
- Chan Yoon Cheah, Sir Charles Gairdner Hospital and PathWest Laboratory Medicine WA, Nedlands; Chan Yoon Cheah, University of Western Australia, Crawley, Western Australia; John F. Seymour, Peter MacCallum Cancer Centre, East Melbourne; John F. Seymour, University of Melbourne, Parkville, Victoria, Australia; and Chan Yoon Cheah and Michael L. Wang, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - John F Seymour
- Chan Yoon Cheah, Sir Charles Gairdner Hospital and PathWest Laboratory Medicine WA, Nedlands; Chan Yoon Cheah, University of Western Australia, Crawley, Western Australia; John F. Seymour, Peter MacCallum Cancer Centre, East Melbourne; John F. Seymour, University of Melbourne, Parkville, Victoria, Australia; and Chan Yoon Cheah and Michael L. Wang, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Michael L Wang
- Chan Yoon Cheah, Sir Charles Gairdner Hospital and PathWest Laboratory Medicine WA, Nedlands; Chan Yoon Cheah, University of Western Australia, Crawley, Western Australia; John F. Seymour, Peter MacCallum Cancer Centre, East Melbourne; John F. Seymour, University of Melbourne, Parkville, Victoria, Australia; and Chan Yoon Cheah and Michael L. Wang, The University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
57
|
Smith SEP, Maus RLG, Davis TR, Sundberg JP, Gil D, Schrum AG. Maternal IL-6 can cause T-cell-mediated juvenile alopecia by non-scarring follicular dystrophy in mice. Exp Dermatol 2015; 25:223-8. [PMID: 26660334 DOI: 10.1111/exd.12914] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2015] [Indexed: 12/16/2022]
Abstract
Aiming to decipher immunological mechanisms of the autoimmune disorder alopecia areata (AA), we hypothesized that interleukin-6 (IL-6) might be associated with juvenile-onset AA, for which there is currently no experimental model. Upon intramuscular transgenesis to overexpress IL-6 in pregnant female C57BL/6 (B6) mice, we found that the offspring displayed an initial normal and complete juvenile hair growth cycle, but developed alopecia around postnatal day 18. This alopecia was patchy and reversible (non-scarring) and was associated with upregulation of Ulbp1 expression, the only mouse homolog of the human AA-associated ULBP3 gene. Alopecia was also associated with inflammatory infiltration of hair follicles by lymphocytes, including alpha-beta T cells, which contributed to surface hair loss. Despite these apparently shared traits with AA, lesions were dominated by follicular dystrophy that was atypical of human AA disease, sharing some traits consistent with B6 alopecia and dermatitis. Additionally, juvenile-onset alopecia was followed by complete, spontaneous recovery of surface hair, without recurrence of hair loss. Prolonging exposure to IL-6 prolonged the time to recovery, but once recovered, repeating high-dose IL-6 exposure de novo did not re-induce alopecia. These data suggest that although substantial molecular and cellular pathways may be shared, functionally similar alopecia disorders can occur via distinct pathological mechanisms.
Collapse
Affiliation(s)
- Stephen E P Smith
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Rachel L G Maus
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Tessa R Davis
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | | | - Diana Gil
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Adam G Schrum
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|
58
|
Chen Z, Teo AE, McCarty N. ROS-Induced CXCR4 Signaling Regulates Mantle Cell Lymphoma (MCL) Cell Survival and Drug Resistance in the Bone Marrow Microenvironment via Autophagy. Clin Cancer Res 2015; 22:187-99. [PMID: 26350264 DOI: 10.1158/1078-0432.ccr-15-0987] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 08/31/2015] [Indexed: 12/12/2022]
Abstract
PURPOSE Patients with advanced stages of mantle cell lymphoma (MCL) have a poor prognosis after standard therapies. MCL cells in those patients often spread into tissues other than lymph nodes, such as the bone marrow. Apart from directed migration and homing, there is little understanding of the function of the CXCR4/SDF-1 signaling axis in MCL. In this report, we aim to understand mechanisms of MCL cell survival in the bone marrow. EXPERIMENTAL DESIGN For comprehensive analyses of MCL interactions with bone marrow stromal cells, we have generated gene knockout cells using CRISPR-CAS9 system and gene knockdown cells to reveal novel roles of the CXCR4/SDF-1 signaling. RESULTS CXCR4 silencing in MCL cells led to a significant reduction in proliferation, cell adhesion to bone marrow stromal cells, and colony formation in PHA-LCM methylcellulose medium, which were reversed upon the addition of SDF-1-neutralizing antibodies. In addition, tracking MCL cell engraftment in vivo revealed that quiescent MCL cells are significantly reduced in the bone marrow upon CXCR4 silencing, indicating that CXCR4/SDF-1 signaling is required for the survival and maintenance of the quiescent MCL cells. Further analysis revealed novel mechanisms of ROS-induced CXCR4/SDF-1 signaling that stimulate autophagy formation in MCL cells for their survival. CONCLUSIONS Our data, for the first time, revealed new roles of the CXCR/SDF-1 signaling axis on autophagy formation in MCL, which further promoted their survival within the bone marrow microenvironment. Targeting the CXCR4/SDF-1/autophagy signaling axis may contribute to an enhanced efficacy of current therapies.
Collapse
Affiliation(s)
- Zheng Chen
- Center for Stem Cell and Regenerative Disease, Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), University of Texas-Health Science Center at Houston, Houston, Texas
| | - Albert E Teo
- Center for Stem Cell and Regenerative Disease, Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), University of Texas-Health Science Center at Houston, Houston, Texas
| | - Nami McCarty
- Center for Stem Cell and Regenerative Disease, Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), University of Texas-Health Science Center at Houston, Houston, Texas.
| |
Collapse
|
59
|
Gribben JG, Fowler N, Morschhauser F. Mechanisms of Action of Lenalidomide in B-Cell Non-Hodgkin Lymphoma. J Clin Oncol 2015; 33:2803-11. [PMID: 26195701 PMCID: PMC5320950 DOI: 10.1200/jco.2014.59.5363] [Citation(s) in RCA: 235] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Lenalidomide is an orally active immunomodulatory drug that has direct antineoplastic activity and indirect effects mediated through multiple types of immune cells found in the tumor microenvironment, including B, T, natural killer (NK), and dendritic cells. Recently, the E3 ubiquitin ligase cereblon was identified as a molecular target that may underlie the effects of lenalidomide on tumor cells, as well as on cells in the tumor microenvironment. Decreases in cereblon attenuate these effects and also confer resistance to lenalidomide. Tumoricidal effects of lenalidomide are associated with reduced interferon regulatory factor 4, a downstream target of cereblon. Lenalidomide stimulates proliferation and activation of NK cells, thereby enhancing NK cell-mediated cytotoxicity and antibody-dependent cellular cytotoxicity. These effects appear to be secondary to cytokine production from T cells. Lenalidomide has been shown to produce synergistic effects in experimental models when evaluated in combination with rituximab, dexamethasone, bortezomib, and B-cell receptor signaling inhibitors, consistent with mechanisms complementary to these agents. These experimental findings have translated to the clinic, where single-agent use displays durable responses in relapsed/refractory non-Hodgkin lymphoma, and combination with rituximab and other agents leads to improved responses at first line and in relapsed/refractory disease. The activity of lenalidomide is evident across multiple lymphoma subtypes, including indolent and aggressive forms. The interaction among cell types in the immune microenvironment is increasingly recognized as important to tumor cell recognition and destruction, as well as to protection of normal immune cells, as reflected by lenalidomide studies across multiple types of B-cell lymphomas.
Collapse
Affiliation(s)
- John G Gribben
- John G. Gribben, Barts Cancer Institute, London, United Kingdom; Nathan Fowler, The University of Texas MD Anderson Cancer Center, Houston, TX; and Franck Morschhauser, Centre Hospitalier Universitaire Régional de Lille, Unité Groupe de Recherche sur les formes Injectables et les Technologies Associées, Lille, France.
| | - Nathan Fowler
- John G. Gribben, Barts Cancer Institute, London, United Kingdom; Nathan Fowler, The University of Texas MD Anderson Cancer Center, Houston, TX; and Franck Morschhauser, Centre Hospitalier Universitaire Régional de Lille, Unité Groupe de Recherche sur les formes Injectables et les Technologies Associées, Lille, France
| | - Franck Morschhauser
- John G. Gribben, Barts Cancer Institute, London, United Kingdom; Nathan Fowler, The University of Texas MD Anderson Cancer Center, Houston, TX; and Franck Morschhauser, Centre Hospitalier Universitaire Régional de Lille, Unité Groupe de Recherche sur les formes Injectables et les Technologies Associées, Lille, France
| |
Collapse
|
60
|
Goobie GC, Bernatsky S, Ramsey-Goldman R, Clarke AE. Malignancies in systemic lupus erythematosus: a 2015 update. Curr Opin Rheumatol 2015; 27:454-60. [PMID: 26125105 PMCID: PMC4562287 DOI: 10.1097/bor.0000000000000202] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Patients with systemic lupus erythematosus (SLE) have altered incidences of certain malignancies as compared with the general population. This review summarizes the recent literature on risk of malignancy in SLE and proposed mechanisms for these altered susceptibilities. RECENT FINDINGS Recent studies have confirmed previous data showing an increased risk of non-Hodgkin's lymphoma, lung, liver, vulvar/vaginal, and thyroid malignancies, whereas demonstrating a decreased risk of breast and prostate cancer. Lymphomagenesis in SLE has been linked to increased activity of multiple inflammatory cytokines as well as possible viral diseases. The decreased rates of hormone-sensitive cancers, such as breast and prostate, are speculated to be related to the presence of lupus autoantibodies and downregulation of certain proteins in SLE. This knowledge has been utilized to investigate new therapeutic modalities for these malignancies. SUMMARY Recent data confirm previously reported altered malignancy rates in SLE. Most striking in recent years are publications further elucidating mechanisms underlying cancer development in SLE, and subsequent investigations of potential therapeutics modulating these pathways.
Collapse
Affiliation(s)
- Gillian C Goobie
- Department of Medicine, Cumming School of Medicine, Health Sciences Centre, Foothills Campus, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, Canada, T2N-4N1. . Phone: 403-710-9242
| | - Sasha Bernatsky
- Departments of Medicine and of Epidemiology and Biostatistics, McGill University, Montreal, Quebec, Canada
| | - Rosalind Ramsey-Goldman
- Department of Medicine, Division of Rheumatology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Ann E. Clarke
- Department of Medicine, Division of Rheumatology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
61
|
Discovery of selective phosphatidylinositol 3-kinase inhibitors to treat hematological malignancies. Drug Discov Today 2015; 20:988-94. [DOI: 10.1016/j.drudis.2015.03.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Revised: 02/22/2015] [Accepted: 03/17/2015] [Indexed: 01/01/2023]
|
62
|
Matsuo K, Hasegawa K, Yoshino K, Murakami R, Hisamatsu T, Stone RL, Previs RA, Hansen JM, Ikeda Y, Miyara A, Hiramatsu K, Enomoto T, Fujiwara K, Matsumura N, Konishi I, Roman LD, Gabra H, Fotopoulou C, Sood AK. Venous thromboembolism, interleukin-6 and survival outcomes in patients with advanced ovarian clear cell carcinoma. Eur J Cancer 2015; 51:1978-88. [PMID: 26238017 DOI: 10.1016/j.ejca.2015.07.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 06/13/2015] [Accepted: 07/13/2015] [Indexed: 01/07/2023]
Abstract
BACKGROUND We compared survival outcomes and risk of venous thromboembolism (VTE) among patients with advanced and early-stage ovarian clear cell carcinoma (OCCC) and serous ovarian carcinoma (SOC), as well as potential links with interleukin-6 (IL-6) levels. METHODS A multicenter case-control study was conducted in 370 patients with OCCC and 938 with SOC. In a subset of 200 cases, pretreatment plasma IL-6 levels were examined. FINDINGS Patients with advanced OCCC had the highest 2-year cumulative VTE rates (advanced OCCC 43.1%, advanced SOC 16.2%, early-stage OCCC 11.9% and early-stage SOC 6.4%, P<0.0001) and the highest median levels of IL-6 (advanced OCCC 17.8 pg/mL, advanced SOC 9.0 pg/mL, early-stage OCCC 4.2 pg/mL and early-stage SOC 5.0 pg/mL, P=0.006). Advanced OCCC (hazard ratio [HR] 3.38, P<0.0001), thrombocytosis (HR 1.42, P=0.032) and elevated IL-6 (HR 8.90, P=0.046) were independent predictors of VTE. In multivariate analysis, patients with advanced OCCC had significantly poorer 5-year progression-free and overall survival rates than those with advanced SOC (P<0.01), and thrombocytosis was an independent predictor of decreased survival outcomes (P<0.01). Elevated IL-6 levels led to poorer 2-year progression-free survival rates in patients with OCCC (50% versus 87.5%, HR 4.89, P=0.016) than in those with SOC (24.9% versus 40.8%, HR 1.40, P=0.07). INTERPRETATION Advanced OCCC is associated with an increased incidence of VTE and decreased survival outcomes, which has major implications for clinical management of OCCC.
Collapse
Affiliation(s)
- Koji Matsuo
- Department of Obstetrics and Gynecology, University of Southern California, Los Angeles, CA, USA; Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA.
| | - Kosei Hasegawa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Kiyoshi Yoshino
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Ryusuke Murakami
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takeshi Hisamatsu
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan; Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rebecca L Stone
- Department of Obstetrics and Gynecology, The Johns Hopkins University, Baltimore, MD, USA
| | - Rebecca A Previs
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jean M Hansen
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yuji Ikeda
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Saitama, Japan; Department of Obstetrics and Gynecology, The University of Tokyo Faculty of Medicine, Tokyo, Japan
| | - Akiko Miyara
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Kosuke Hiramatsu
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takayuki Enomoto
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medicine, Niigata, Japan
| | - Keiichi Fujiwara
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Noriomi Matsumura
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ikuo Konishi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Lynda D Roman
- Department of Obstetrics and Gynecology, University of Southern California, Los Angeles, CA, USA; Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Hani Gabra
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Christina Fotopoulou
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Anil K Sood
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
63
|
Lu K, Chen N, Zhou XX, Ge XL, Feng LL, Li PP, Li XY, Geng LY, Wang X. The STAT3 inhibitor WP1066 synergizes with vorinostat to induce apoptosis of mantle cell lymphoma cells. Biochem Biophys Res Commun 2015; 464:292-8. [PMID: 26116769 DOI: 10.1016/j.bbrc.2015.06.145] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 06/22/2015] [Indexed: 12/19/2022]
Abstract
Mantle cell lymphoma (MCL) is an aggressive B-cell non-Hodgkin lymphoma (NHL) characterized by the translocation t (11; 14) (q13; q32). Drug resistance remains a formidable obstacle to treatment and the median survival for MCL patients is between 3 and 5 years. Thus, there is an urgent need to discover novel approaches to MCL therapy. The signal transducer and activation of transcription 3 (STAT3) has been found to be constitutively activated in several subtypes of MCL cell lines and MCL tumors. WP1066, a small-molecule inhibitor of STAT3, exerted antitumor activity in hematological and solid malignancies by inhibiting key survival and growth signaling pathways. In the present study, we evaluated the antiproliferative and proapoptotic activity of WP1066 combined with pan-histone deacetylase (HDAC) inhibitor vorinostat (SAHA) in a panel of MCL cell lines. In addition, potential mechanisms involved were also explored. The outcome showed that combination of WP1066 with SAHA resulted in synergistic growth inhibition and apoptosis induction in MCL cell lines in vitro. Furthermore, combination of WP1066 with SAHA inhibited the constitutive STAT3 activation and modulated mRNA expressions of anti- and pro-apoptotic genes. Our findings suggest that agents targeting the STAT3 pathway such as WP1066 may be useful therapeutic drugs for MCL when combined with SAHA.
Collapse
Affiliation(s)
- Kang Lu
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, PR China
| | - Na Chen
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, PR China
| | - Xiang-xiang Zhou
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, PR China
| | - Xue-ling Ge
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, PR China
| | - Li-li Feng
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, PR China
| | - Pei-pei Li
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, PR China
| | - Xin-yu Li
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, PR China
| | - Ling-yun Geng
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, PR China
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, PR China; Institute of Diagnostics, Shandong University School of Medicine, Jinan, Shandong 250021, PR China.
| |
Collapse
|
64
|
Tucker DL, Rule SA. A critical appraisal of ibrutinib in the treatment of mantle cell lymphoma and chronic lymphocytic leukemia. Ther Clin Risk Manag 2015; 11:979-90. [PMID: 26150724 PMCID: PMC4484687 DOI: 10.2147/tcrm.s73559] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Although chemo-immunotherapy remains at the forefront of first-line treatment for mantle cell lymphoma (MCL) and chronic lymphocytic leukemia (CLL), small molecules, such as ibrutinib, are beginning to play a significant role, particularly in patients with multiply relapsed or chemotherapy-refractory disease and where toxicity is an overriding concern. Ibrutinib is a first-in-class, oral inhibitor of Bruton's tyrosine kinase, which functions by irreversible inhibition of the downstream signaling pathway of the B-cell receptor, which normally promotes cell survival and proliferation. Early clinical trials have demonstrated excellent tolerability and a modest side-effect profile even in elderly and multiply pretreated patient cohorts. Although the majority of disease responses tend to be partial, efficacy data have also been encouraging with more than two-thirds of patients with CLL and MCL demonstrating a durable response, even in the high-risk disease setting. Resistance mechanisms are only partially understood and appear to be multifactorial, including the binding site mutation C481S, and escape through other common cell-signaling pathways. This article appraises the currently available data on safety and efficacy from clinical trials of ibrutinib in the management of MCL and CLL, both as a single agent and in combination with other therapies, and considers how this drug is likely to be used in future clinical practice.
Collapse
Affiliation(s)
- David L Tucker
- Department of Haematology, Plymouth Hospitals NHS Trust, Plymouth, UK
| | - Simon A Rule
- Department of Haematology, Plymouth Hospitals NHS Trust, Plymouth, UK
| |
Collapse
|
65
|
Dual PI3K/mTOR inhibition is required to effectively impair microenvironment survival signals in mantle cell lymphoma. Oncotarget 2015; 5:6788-800. [PMID: 25216518 PMCID: PMC4196163 DOI: 10.18632/oncotarget.2253] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Phosphatidylinositol-3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway activation contributes to mantle cell lymphoma (MCL) pathogenesis and drug resistance. Antitumor activity has been observed with mTOR inhibitors. However, they have shown limited clinical efficacy in relation to drug activation of feedback loops. Selective PI3K inhibition or dual PI3K/mTOR catalytic inhibition are different therapeutic approaches developed to achieve effective pathway blockage. Here, we have performed a comparative analysis of the mTOR inhibitor everolimus, the pan-PI3K inhibitor NVP-BKM120 and the dual PI3K/mTOR inhibitor NVP-BEZ235 in primary MCL cells. We found NVP-BEZ235 to be more powerful than everolimus or NVP-BKM120 in PI3K/Akt/mTOR signaling inhibition, indicating that targeting the PI3K/Akt/mTOR pathway at multiple levels is likely to be a more effective strategy for the treatment of MCL than single inhibition of these kinases. Among the three drugs, NVP-BEZ235 induced the highest change in gene expression profile. Functional validation demonstrated that NVP-BEZ235 inhibited angiogenesis, migration and tumor invasiveness in MCL cells. NVP-BEZ235 was the only drug able to block IL4 and IL6/STAT3 signaling which compromise the therapeutic effect of chemotherapy in MCL. Our findings support the use of the dual PI3K/mTOR inhibitor NVP-BEZ235 as a promising approach to interfere with the microenvironment-related processes in MCL.
Collapse
|
66
|
Differential PAX5 levels promote malignant B-cell infiltration, progression and drug resistance, and predict a poor prognosis in MCL patients independent of CCND1. Leukemia 2015; 30:580-93. [PMID: 26073757 PMCID: PMC4644730 DOI: 10.1038/leu.2015.140] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 05/06/2015] [Accepted: 05/11/2015] [Indexed: 01/06/2023]
Abstract
Reduced Paired box 5 (PAX5) levels have important roles in the pathogenesis of human B-cell acute lymphoblastic leukemia. However, the role of PAX5 in human lymphoma remains unclear. We generated PAX5-silenced cells using mantle cell lymphoma (MCL) as a model system. These PAX5(-) MCL cells exhibited unexpected phenotypes, including increased proliferation in vitro, enhanced tumor infiltration in vivo, robust adhesion to the bone marrow stromal cells and increased retention of quiescent stem-like cells. These phenotypes were attributed to alterations in the expression of genes including p53 and Rb, and to phosphoinositide 3-kinase/mammalian target of rapamycin and phosphorylated signal transducer and activator of transcription 3 pathway hyperactivation. On PAX5 silencing, the MCL cells displayed upregulated interleukin (IL)-6 expression and increased responses to paracrine IL-6. Moreover, decreased PAX5 levels in CD19+ MCL cells correlated with their increased infiltration and progression; thus, PAX5 levels can be used as a prognostic marker independent of cyclin D1 in advanced MCL patients. Importantly, high-throughput screening of 3800 chemical compounds revealed that PAX5(-) MCL cells are highly drug-resistant compared with PAX5 wild-type MCL cells. Collectively, the results of our study support a paradigm shift regarding the functions of PAX5 in human B-cell cancer and encourage future efforts to design effective therapies against MCL.
Collapse
|
67
|
Tang J, Zhu J, Yu Y, Zhang Z, Chen G, Zhou X, Qiao C, Hou T, Mao X. A virtual screen identified C96 as a novel inhibitor of phosphatidylinositol 3-kinase that displays potent preclinical activity against multiple myeloma in vitro and in vivo. Oncotarget 2015; 5:3836-48. [PMID: 25003534 PMCID: PMC4116524 DOI: 10.18632/oncotarget.1657] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The phosphatidylinositol 3-kinase (PI3K)/AKT signaling pathway is emerging as a promising therapeutic target for multiple myeloma (MM). In the present study, we performed a virtual screen against 800,000 of small molecule compounds by targeting PI3Kγ. C96, one of such compounds, inhibited PI3K activated by insulin-like growth factor-1 (IGF-1), but did not suppress IGF-1R activation. The cell-free assay revealed that C96 preferred to inhibit PI3Kα and δ, but was not active against AKT1, 2, 3 or mTOR. C96 inhibited PI3K activation in a time- and concentration-dependent manner. Consistent with its inhibition on PI3K/AKT, C96 downregulated the activation of mTOR, p70S6K, 4E-BP1, but did not suppress other kinases such as ERK and c-Src. Inhibition of the PI3K/AKT signaling pathway by C96 led to MM cell apoptosis which was demonstrated by Annexin V staining and activation of the pro-apoptotic signals. Furthermore, C96 displayed potent anti-myeloma activity in a MM xenograft model in nude mice. Oral administration of 100 mg/kg bodyweight almost fully suppressed tumor growth within 16 days, but without gross toxicity. Importantly, AKT activation was suppressed in tumor tissues from C96-treated mice, which was consistent with delayed tumor growth. Thus, we identified a novel PI3K inhibitor with a great potential for MM therapy.
Collapse
Affiliation(s)
- Juan Tang
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China;Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital, Soochow University, Suzhou, China
| | | | | | | | | | | | | | | | - Xinliang Mao
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China; Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital, Soochow University, Suzhou, China;Collaborative Innovation Center of Hematology, Suzhou, China
| |
Collapse
|
68
|
Crescenzo R, Abate F, Lasorsa E, Tabbo' F, Gaudiano M, Chiesa N, Di Giacomo F, Spaccarotella E, Barbarossa L, Ercole E, Todaro M, Boi M, Acquaviva A, Ficarra E, Novero D, Rinaldi A, Tousseyn T, Rosenwald A, Kenner L, Cerroni L, Tzankov A, Ponzoni M, Paulli M, Weisenburger D, Chan WC, Iqbal J, Piris MA, Zamo' A, Ciardullo C, Rossi D, Gaidano G, Pileri S, Tiacci E, Falini B, Shultz LD, Mevellec L, Vialard JE, Piva R, Bertoni F, Rabadan R, Inghirami G. Convergent mutations and kinase fusions lead to oncogenic STAT3 activation in anaplastic large cell lymphoma. Cancer Cell 2015; 27:516-32. [PMID: 25873174 PMCID: PMC5898430 DOI: 10.1016/j.ccell.2015.03.006] [Citation(s) in RCA: 348] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 11/14/2014] [Accepted: 03/10/2015] [Indexed: 01/01/2023]
Abstract
A systematic characterization of the genetic alterations driving ALCLs has not been performed. By integrating massive sequencing strategies, we provide a comprehensive characterization of driver genetic alterations (somatic point mutations, copy number alterations, and gene fusions) in ALK(-) ALCLs. We identified activating mutations of JAK1 and/or STAT3 genes in ∼20% of 88 [corrected] ALK(-) ALCLs and demonstrated that 38% of systemic ALK(-) ALCLs displayed double lesions. Recurrent chimeras combining a transcription factor (NFkB2 or NCOR2) with a tyrosine kinase (ROS1 or TYK2) were also discovered in WT JAK1/STAT3 ALK(-) ALCL. All these aberrations lead to the constitutive activation of the JAK/STAT3 pathway, which was proved oncogenic. Consistently, JAK/STAT3 pathway inhibition impaired cell growth in vitro and in vivo.
Collapse
Affiliation(s)
- Ramona Crescenzo
- Department of Molecular Biotechnology and Health Science and Center for Experimental Research and Medical Studies, University of Torino, 10126 Torino, Italy; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Francesco Abate
- Department of Molecular Biotechnology and Health Science and Center for Experimental Research and Medical Studies, University of Torino, 10126 Torino, Italy; Department of Control and Computer Engineering, Politecnico di Torino, 10129 Torino, Italy; Department of Biomedical Informatics and Department of Systems Biology, Center for Computational Biology and Bioinformatics, Columbia University, New York, NY 10027, USA
| | - Elena Lasorsa
- Department of Molecular Biotechnology and Health Science and Center for Experimental Research and Medical Studies, University of Torino, 10126 Torino, Italy
| | - Fabrizio Tabbo'
- Department of Molecular Biotechnology and Health Science and Center for Experimental Research and Medical Studies, University of Torino, 10126 Torino, Italy; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Marcello Gaudiano
- Department of Molecular Biotechnology and Health Science and Center for Experimental Research and Medical Studies, University of Torino, 10126 Torino, Italy; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Nicoletta Chiesa
- Department of Molecular Biotechnology and Health Science and Center for Experimental Research and Medical Studies, University of Torino, 10126 Torino, Italy
| | - Filomena Di Giacomo
- Department of Molecular Biotechnology and Health Science and Center for Experimental Research and Medical Studies, University of Torino, 10126 Torino, Italy
| | - Elisa Spaccarotella
- Department of Molecular Biotechnology and Health Science and Center for Experimental Research and Medical Studies, University of Torino, 10126 Torino, Italy
| | - Luigi Barbarossa
- Department of Molecular Biotechnology and Health Science and Center for Experimental Research and Medical Studies, University of Torino, 10126 Torino, Italy
| | - Elisabetta Ercole
- Department of Molecular Biotechnology and Health Science and Center for Experimental Research and Medical Studies, University of Torino, 10126 Torino, Italy
| | - Maria Todaro
- Department of Molecular Biotechnology and Health Science and Center for Experimental Research and Medical Studies, University of Torino, 10126 Torino, Italy; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Michela Boi
- Department of Molecular Biotechnology and Health Science and Center for Experimental Research and Medical Studies, University of Torino, 10126 Torino, Italy; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Andrea Acquaviva
- Department of Control and Computer Engineering, Politecnico di Torino, 10129 Torino, Italy
| | - Elisa Ficarra
- Department of Control and Computer Engineering, Politecnico di Torino, 10129 Torino, Italy
| | - Domenico Novero
- Department of Pathology, A.O. Città della Salute e della Scienza (Molinette), 10126 Torino, Italy
| | - Andrea Rinaldi
- Lymphoma and Genomics Research Program, Institute of Oncology Research, 6500 Bellinzona, Switzerland
| | - Thomas Tousseyn
- Translational Cell and Tissue Research Lab, KU Leuven, 3000 Leuven, Belgium
| | - Andreas Rosenwald
- Institute of Pathology, University of Würzburg and Comprehensive Cancer Center Mainfranken, 97080 Würzburg, Germany
| | - Lukas Kenner
- Ludwing Boltzmann Institute for Cancer Research, 1090 Vienna, Austria
| | - Lorenzo Cerroni
- Research Unit Dermatopathology of the Medical University of Graz, 8036 Graz, Austria
| | - Alexander Tzankov
- Institute of Pathology, University Hospital Basel, 4031 Basel, Switzerland
| | - Maurilio Ponzoni
- Pathology & Lymphoid Malignancies Units, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Marco Paulli
- Department of Human Pathology, University of Pavia and Scientific Institute Fondazione Policlinico San Matteo, 27100 Pavia, Italy
| | | | - Wing C Chan
- Department of Pathology, City of Hope Medical Center, Duarte, CA 91010, USA
| | - Javeed Iqbal
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Miguel A Piris
- Cancer Genomics, Instituto de Formación e Investigación Marqués de Valdecilla and Department of Pathology, Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain
| | - Alberto Zamo'
- Department of Pathology and Diagnostics, University of Verona, 37134 Verona, Italy
| | - Carmela Ciardullo
- Division of Hematology, Department of Translational Medicine, Amedeo Avogadro University of Eastern Piedmont, 28100 Novara, Italy
| | - Davide Rossi
- Division of Hematology, Department of Translational Medicine, Amedeo Avogadro University of Eastern Piedmont, 28100 Novara, Italy
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, Amedeo Avogadro University of Eastern Piedmont, 28100 Novara, Italy
| | - Stefano Pileri
- European Institute of Oncology, 20141 Milano, Italy; Bologna University School of Medicine, 40126 Bologna, Italy
| | - Enrico Tiacci
- Institute of Hematology-Centro di Ricerche Onco-Ematologiche (CREO), Ospedale S. Maria della Misericordia, University of Perugia, 06100 Perugia, Italy
| | - Brunangelo Falini
- Institute of Hematology-Centro di Ricerche Onco-Ematologiche (CREO), Ospedale S. Maria della Misericordia, University of Perugia, 06100 Perugia, Italy
| | | | - Laurence Mevellec
- Janssen Research & Development, a Division of Janssen-Cilag, Campus de Maigremont, CS10615, 27106 Val-de-Reuil Cedex, France
| | - Jorge E Vialard
- Janssen Research & Development, a Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Roberto Piva
- Department of Molecular Biotechnology and Health Science and Center for Experimental Research and Medical Studies, University of Torino, 10126 Torino, Italy; Department of Pathology and NYU Cancer Center, New York University School of Medicine, New York, NY 10016, USA
| | - Francesco Bertoni
- Lymphoma and Genomics Research Program, Institute of Oncology Research, 6500 Bellinzona, Switzerland; Oncology Institute of Southern Switzerland, 6500 Bellinzona, Switzerland
| | - Raul Rabadan
- Department of Biomedical Informatics and Department of Systems Biology, Center for Computational Biology and Bioinformatics, Columbia University, New York, NY 10027, USA.
| | - Giorgio Inghirami
- Department of Molecular Biotechnology and Health Science and Center for Experimental Research and Medical Studies, University of Torino, 10126 Torino, Italy; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY 10021, USA; Department of Pathology and NYU Cancer Center, New York University School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
69
|
Blonska M, Agarwal NK, Vega F. Shaping of the tumor microenvironment: Stromal cells and vessels. Semin Cancer Biol 2015; 34:3-13. [PMID: 25794825 DOI: 10.1016/j.semcancer.2015.03.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Revised: 03/04/2015] [Accepted: 03/08/2015] [Indexed: 12/12/2022]
Abstract
Lymphomas develop and progress in a specialized tissue microenvironment such as bone marrow as well as secondary lymphoid organs such as lymph node and spleen. The lymphoma microenvironment is characterized by a heterogeneous population of stromal cells, including fibroblastic reticular cells, nurse-like cells, mesenchymal stem cells, follicular dendritic cells, and inflammatory cells such as macrophages, T- and B-cells. These cell populations interact with the lymphoma cells to promote lymphoma growth, survival and drug resistance through multiple mechanisms. Angiogenesis is also recognized as an important factor associated with lymphoma progression. In recent years, we have learned that the interaction between the malignant and non-malignant cells is bidirectional and resembles, at least in part, the pattern seen between non-neoplastic lymphoid cells and the normal microenvironment of lymphoid organs. A summary of the current knowledge of lymphoma microenvironment focusing on the cellular components will be reviewed here.
Collapse
Affiliation(s)
- Marzenna Blonska
- Division of Hematology-Oncology, Department of Medicine, University of Miami and Sylvester Comprehensive Cancer Center, Miami, FL, United States
| | - Nitin K Agarwal
- Division of Hematopathology, Department of Pathology, University of Miami and Sylvester Comprehensive Cancer Center, Miami, FL, United States
| | - Francisco Vega
- Division of Hematopathology, Department of Pathology, University of Miami and Sylvester Comprehensive Cancer Center, Miami, FL, United States.
| |
Collapse
|
70
|
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: 8.0] [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
|
71
|
Zhang N, Li YX, Tao L, Yang L, Zhao J, Zhang WJ. Clinicopathological and prognostic significance of IL-11 and Survivin overexpression in gastric cancer. Shijie Huaren Xiaohua Zazhi 2015; 23:373-380. [DOI: 10.11569/wcjd.v23.i3.373] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the differential expression of interleukin-11 (IL-11) and Survivin in gastric cancer (GC) to understand their possible roles in carcinogenesis, progression and prognosis.
METHODS: Paraffin-embedded samples were obtained from 59 GC patients who underwent surgical operations at the First Affiliated Hospital of Shihezi University School of Medicine, Shihezi, China, between January 1, 2004 and December 31, 2007. All of the patients were followed to April 1, 20014. The expression of IL-11 and Survivin was detected using tissue chip and immunohistochemistry.
RESULTS: The positive expression rate of IL-11 was 96.6% in GC tissues, significantly higher than that in adjacent normal tissues (88.1%; χ2 = 7.252, (P = 0.025). The positive expression rate of Survivin in GC tissues was also significantly higher than that in adjacent normal tissues (93.2% vs 72.9%, χ2 = 41.988, P < 0.001). In GC tissues, there was a positive correlation between IL-11 and Survivin expression (r = 0.442, P < 0.001), but no correlation was found in adjacent normal tissues (r = 0.103, (P = 0.438). The expression of IL-11 was correlated with higher clinical stage ((P = 0.002), while Survivin expression was correlated with lymph node metastasis and higher clinical stage ((P = 0.002, 0.028). Higher levels of IL-11 and Survivin were linked to poor prognosis in GC patients ((P = 0.004 and P < 0.001, respectively). Cox multi-factorial regression analysis demonstrated clinical stage being an independent factor predicting overall survival of GC patients ((P = 0.017).
CONCLUSION: Overexpression of IL-11 and Survivin correlates with poor prognosis of GC patients, and they may play a coordinated role in the development, progression and prognosis of GC.
Collapse
|
72
|
Abstract
Human IL6 is a cytokine produced by many cell types that has pleiotropic effects. In agreement, anti-IL6 therapy reduces inflammation, hepatic acute phase proteins, and anemia and has antiangiogenic effects. Blocking IL6 has demonstrated therapeutic efficacy with drug registration in Castleman disease and inflammatory diseases (rheumatoid arthritis) without major toxicity. Interestingly, the inhibition of C-reactive protein (CRP) production is a trustworthy surrogate marker of anti-IL6 therapy efficacy. Clinically registered IL6 inhibitors include siltuximab, an anti-IL6 mAb, and tocilizumab, an anti-IL6R mAb. In various cancers, in particular plasma cell cancers, large randomized trials showed no efficacy of IL6 inhibitors, despite a full inhibition of CRP production in treated patients in vivo, the numerous data showing an involvement of IL6 in these diseases, and initial short-term treatments demonstrating a dramatic inhibition of cancer cell proliferation in vivo. A likely explanation is the plasticity of cancer cells, with the presence of various subclones, making the outgrowth of cancer subclones possible using growth factors other than IL6. In addition, current therapeutic strategies used in these cancers already target IL6 activity. Thus, anti-IL6 therapeutics are able to neutralize IL6 production in vivo and are safe and useful in inflammatory diseases and Castleman disease.
Collapse
Affiliation(s)
- Jean-François Rossi
- Department of Hematology, CHU de Montpellier, Montpellier, France. INSERM U1040, Montpellier, France. Université Montpellier I, Montpellier, France.
| | - Zhao-Yang Lu
- Unité de Thérapie Cellulaire, CHU de Montpellier, Montpellier, France
| | | | - Bernard Klein
- INSERM U1040, Montpellier, France. Université Montpellier I, Montpellier, France
| |
Collapse
|
73
|
Yu H, Lee H, Herrmann A, Buettner R, Jove R. Revisiting STAT3 signalling in cancer: new and unexpected biological functions. Nat Rev Cancer 2014; 14:736-46. [PMID: 25342631 DOI: 10.1038/nrc3818] [Citation(s) in RCA: 1636] [Impact Index Per Article: 148.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The Janus kinases (JAKs) and signal transducer and activator of transcription (STAT) proteins, particularly STAT3, are among the most promising new targets for cancer therapy. In addition to interleukin-6 (IL-6) and its family members, multiple pathways, including G-protein-coupled receptors (GPCRs), Toll-like receptors (TLRs) and microRNAs were recently identified to regulate JAK-STAT signalling in cancer. Well known for its role in tumour cell proliferation, survival, invasion and immunosuppression, JAK-STAT3 signalling also promotes cancer through inflammation, obesity, stem cells and the pre-metastatic niche. In addition to its established role as a transcription factor in cancer, STAT3 regulates mitochondrion functions, as well as gene expression through epigenetic mechanisms. Newly identified regulators and functions of JAK-STAT3 in tumours are important targets for potential therapeutic strategies in the treatment of cancer.
Collapse
Affiliation(s)
- Hua Yu
- Department of Cancer Immunotherapeutics and Tumor Immunology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California 91010, USA
| | - Heehyoung Lee
- Department of Cancer Immunotherapeutics and Tumor Immunology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California 91010, USA
| | - Andreas Herrmann
- Department of Cancer Immunotherapeutics and Tumor Immunology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California 91010, USA
| | - Ralf Buettner
- Department of Molecular Medicine, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California 91010, USA
| | - Richard Jove
- Vaccine and Gene Therapy Institute of Florida, Port St. Lucie, Florida 34987, USA
| |
Collapse
|
74
|
Hong B, Li H, Zhang M, Xu J, Lu Y, Zheng Y, Qian J, Chang JT, Yang J, Yi Q. p38 MAPK inhibits breast cancer metastasis through regulation of stromal expansion. Int J Cancer 2014; 136:34-43. [PMID: 24806617 DOI: 10.1002/ijc.28958] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 04/25/2014] [Indexed: 01/14/2023]
Abstract
p38 MAPK signaling controls cell growth, proliferation and the cell cycle under stress conditions. However, the function of p38 activation in tumor metastasis is still not well understood. We report that p38 activation in breast cancer cells inhibits tumor metastasis but does not substantially modulate primary tumor growth. Stable p38 knockdown in breast cancer cells suppressed NF-κB p65 activation, inhibiting miR-365 expression and resulting in increased IL-6 secretion. The inhibitory effect of p38 signaling on metastasis was mediated by suppression of mesenchymal stem cell (MSC) migration to the primary tumor and sites of metastasis, where MSCs can differentiate into cancer-associated fibroblasts to promote tumor metastasis. The migration of MSCs to these sites relies on CXCR4-SDF1 signaling in the tumor microenvironment. Analysis of human primary and metastatic breast cancer tumors showed that p38 activation was inversely associated with IL-6 and vimentin expression. This study suggests that combination analysis of p38 MAPK and IL-6 signaling in patients with breast cancer may improve prognosis and treatment of metastatic breast cancer.
Collapse
Affiliation(s)
- Bangxing Hong
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Nakayama S, Yokote T, Tsuji M, Hirata Y, Iwaki K, Takayama A, Nishiwaki U, Hiraoka N, Akioka T, Miyoshi T, Masuda Y, Hanafusa T. Multiple cytokine-producing testicular malignant lymphoma with clinical symptoms resembling infectious signs. Int J Immunopathol Pharmacol 2014; 27:103-7. [PMID: 24674684 DOI: 10.1177/039463201402700113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
We present the case of a 64-year-old male with painful swelling of the bilateral testes and epididymides, high fever, leukocytosis, and an elevated C-reactive protein (CRP) level. This is the first case report of testicular diffuse large B-cell lymphoma, not otherwise specified (DLBCL, NOS) immunostained for multiple cytokines and their receptors, which clearly demonstrates that tumor cells express multiple cytokines [interleukin-6 (IL-6) and granulocyte colony-stimulating factor (G-CSF)] and their receptors [IL-6 receptor (IL-6R) and G-CSF receptor (G-CSFR)]. The clinical course showed that the reduction in tumor size was accompanied by a corresponding improvement in clinical symptoms and peripheral blood findings. Such clinical investigation may lead clinicians to misdiagnose inflammatory disease rather than neoplastic disease. Recognizing this paraneoplastic phenomenon associated with some cases of testicular DLBCL, NOS is important. In addition, this case suggests that the growth of tumor cells may be promoted through autocrine mechanisms of IL-6 and G-CSF, which are produced by tumor cells. The possibility that these cytokines can be produced by tumor cells and can accelerate tumor proliferation should be considered to be a cause of severe clinical symptoms, an aggressive clinical course, and an indication of the necessity of treatment. Certain cytokines may be used as tumor markers in some cases of DLBCL, NOS.
Collapse
Affiliation(s)
- S Nakayama
- Department of Internal Medicine (I), Osaka Medical College, Japan
| | - T Yokote
- Department of Internal Medicine (I), Osaka Medical College, Japan
| | - M Tsuji
- Division of Surgical Pathology, Osaka Medical College, Osaka, Japan
| | - Y Hirata
- Department of Internal Medicine (I), Osaka Medical College, Japan
| | - K Iwaki
- Department of Internal Medicine (I), Osaka Medical College, Japan
| | - A Takayama
- Department of Internal Medicine (I), Osaka Medical College, Japan
| | - U Nishiwaki
- Department of Internal Medicine (I), Osaka Medical College, Japan
| | - N Hiraoka
- Department of Internal Medicine (I), Osaka Medical College, Japan
| | - T Akioka
- Department of Internal Medicine (I), Osaka Medical College, Japan
| | - T Miyoshi
- Department of Internal Medicine (I), Osaka Medical College, Japan
| | - Y Masuda
- Department of Internal Medicine (I), Osaka Medical College, Japan
| | - T Hanafusa
- Department of Internal Medicine (I), Osaka Medical College, Japan
| |
Collapse
|
76
|
Yao J, Qian CJ, Ye B, Zhao ZQ, Wei J, Liang Y, Zhang X. Signal transducer and activator of transcription 3 signaling upregulates fascin via nuclear factor-κB in gastric cancer: Implications in cell invasion and migration. Oncol Lett 2014; 7:902-908. [PMID: 24527098 PMCID: PMC3919871 DOI: 10.3892/ol.2014.1804] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 12/20/2013] [Indexed: 12/13/2022] Open
Abstract
Fascin protein plays important roles in tumor metastasis and is prognostically relevant to human gastric cancer (GC). However, its role in the development and progression of GC has not been comprehensively investigated. In the present study, results revealed that upregulation of fascin by interleukin-6 promotes GC cell migration and invasion in a signal transducer and activator of transcription 3 (STAT3)-dependent manner in MKN45 cells. Furthermore, STAT3 directly regulated fascin expression and nuclear factor-κB (NF-κB) bound to the fascin promoter in a STAT3-dependent and Notch-independent manner. Therefore, results demonstrate that STAT3 and NF-κB are required for upregulation of fascin and for cell migration and invasion in MKN45 cells. Effects of the treatments on cell signaling were detected by qPCR, western blot analysis and chromatin immunoprecipitation (ChIP) assay. Cell migration and invasion were analyzed using in vitro scratch wound healing assay, transwell and Matrigel assays, and xenograft model. In addition, the STAT3-NF-κB-fascin signaling axis is identified as a therapeutic target for blocking GC cell invasion and migration.
Collapse
Affiliation(s)
- Jun Yao
- Institute of Tumor, School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| | - Cui-Juan Qian
- Institute of Tumor, School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, P.R. China ; Insitute of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Bei Ye
- Department of Gastroenterology, Taizhou Municipal Hospital, Taizhou, Zhejiang 318000, P.R. China
| | - Zhi-Qiang Zhao
- Institute of Tumor, School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| | - Jie Wei
- Institute of Tumor, School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| | - Yong Liang
- Institute of Tumor, School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| | - Xin Zhang
- Department of Gastroenterology, Taizhou Municipal Hospital, Taizhou, Zhejiang 318000, P.R. China
| |
Collapse
|
77
|
Qian C, Yao J, Wang J, Wang L, Xue M, Zhou T, Liu W, Si J. ERK1/2 inhibition enhances apoptosis induced by JAK2 silencing in human gastric cancer SGC7901 cells. Mol Cell Biochem 2013; 387:159-70. [PMID: 24178240 DOI: 10.1007/s11010-013-1881-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2013] [Accepted: 10/18/2013] [Indexed: 12/11/2022]
Abstract
Recent studies suggest JAK2 signaling may be a therapeutic target for treatment of gastric cancer (GC). However, the exact roles of JAK2 in gastric carcinogenesis are not very clear. Here, we have targeted JAK2 to be silenced by shRNA and investigated the biological functions and related mechanisms of JAK2 in GC cell SGC7901. In this study, JAK2 is commonly highly expressed in GC tissues as compared to their adjacent normal tissues (n = 75, p < 0.01). Specific down-regulation of JAK2 suppressed cell proliferation and colony-forming units, induced G2/M arrest in SGC7901 cells, but had no significant effect on cell apoptosis in vitro or tumor growth inhibition in vivo. Interestingly, JAK2 silencing-induced activation of ERK1/2, and inactivation of ERK1/2 using the specific ERK inhibitor PD98059 markedly enhanced JAK2 shRNA-induced cell proliferation inhibition, cell cycle arrest and apoptosis. Ultimately, combination of PD98059 and JAK2 shRNA significantly inhibited tumor growth in nude mice. Our results implicate JAK2 silencing-induced cell proliferation inhibition, cell cycle arrest, and ERK1/2 inhibition could enhance apoptosis induced by JAK2 silencing in SGC7901 cells.
Collapse
Affiliation(s)
- Cuijuan Qian
- Institute of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, Zhejiang, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
78
|
Landscape of somatic mutations and clonal evolution in mantle cell lymphoma. Proc Natl Acad Sci U S A 2013; 110:18250-5. [PMID: 24145436 DOI: 10.1073/pnas.1314608110] [Citation(s) in RCA: 407] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Mantle cell lymphoma (MCL) is an aggressive tumor, but a subset of patients may follow an indolent clinical course. To understand the mechanisms underlying this biological heterogeneity, we performed whole-genome and/or whole-exome sequencing on 29 MCL cases and their respective matched normal DNA, as well as 6 MCL cell lines. Recurrently mutated genes were investigated by targeted sequencing in an independent cohort of 172 MCL patients. We identified 25 significantly mutated genes, including known drivers such as ataxia-telangectasia mutated (ATM), cyclin D1 (CCND1), and the tumor suppressor TP53; mutated genes encoding the anti-apoptotic protein BIRC3 and Toll-like receptor 2 (TLR2); and the chromatin modifiers WHSC1, MLL2, and MEF2B. We also found NOTCH2 mutations as an alternative phenomenon to NOTCH1 mutations in aggressive tumors with a dismal prognosis. Analysis of two simultaneous or subsequent MCL samples by whole-genome/whole-exome (n = 8) or targeted (n = 19) sequencing revealed subclonal heterogeneity at diagnosis in samples from different topographic sites and modulation of the initial mutational profile at the progression of the disease. Some mutations were predominantly clonal or subclonal, indicating an early or late event in tumor evolution, respectively. Our study identifies molecular mechanisms contributing to MCL pathogenesis and offers potential targets for therapeutic intervention.
Collapse
|
79
|
|
80
|
Manni S, Brancalion A, Mandato E, Tubi LQ, Colpo A, Pizzi M, Cappellesso R, Zaffino F, Di Maggio SA, Cabrelle A, Marino F, Zambello R, Trentin L, Adami F, Gurrieri C, Semenzato G, Piazza F. Protein kinase CK2 inhibition down modulates the NF-κB and STAT3 survival pathways, enhances the cellular proteotoxic stress and synergistically boosts the cytotoxic effect of bortezomib on multiple myeloma and mantle cell lymphoma cells. PLoS One 2013; 8:e75280. [PMID: 24086494 PMCID: PMC3785505 DOI: 10.1371/journal.pone.0075280] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 08/14/2013] [Indexed: 02/07/2023] Open
Abstract
CK2 is a pivotal pro-survival protein kinase in multiple myeloma that may likely impinge on bortezomib-regulated cellular pathways. In the present study, we investigated CK2 expression in multiple myeloma and mantle cell lymphoma, two bortezomib-responsive B cell tumors, as well as its involvement in bortezomib-induced cytotoxicity and signaling cascades potentially mediating bortezomib resistance. In both tumors, CK2 expression correlated with that of its activated targets NF-κB and STAT3 transcription factors. Bortezomib-induced proliferation arrest and apoptosis were significantly amplified by the simultaneous inhibition of CK2 with two inhibitors (CX-4945 and K27) in multiple myeloma and mantle cell lymphoma cell lines, in a model of multiple myeloma bone marrow microenvironment and in cells isolated from patients. CK2 inhibition empowered bortezomib-triggered mitochondrial-dependent cell death. Phosphorylation of NF-κB p65 on Ser529 (a CK2 target site) and rise of the levels of the endoplasmic reticulum stress kinase/endoribonuclease Ire1α were markedly reduced upon CK2 inhibition, as were STAT3 phospho Ser727 levels. On the contrary, CK2 inhibition increased phospho Ser51 eIF2α levels and enhanced the bortezomib-dependent accumulation of poly-ubiquitylated proteins and of the proteotoxic stress-associated chaperone Hsp70. Our data suggest that CK2 over expression in multiple myeloma and mantle cell lymphoma cells might sustain survival signaling cascades and can antagonize bortezomib-induced apoptosis at different levels. CK2 inhibitors could be useful in bortezomib-based combination therapies.
Collapse
Affiliation(s)
- Sabrina Manni
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Laboratory, Hematologic Malignancies Unit, Venetian Institute of Molecular Medicine, Padova, Italy
| | - Alessandra Brancalion
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Laboratory, Hematologic Malignancies Unit, Venetian Institute of Molecular Medicine, Padova, Italy
| | - Elisa Mandato
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Laboratory, Hematologic Malignancies Unit, Venetian Institute of Molecular Medicine, Padova, Italy
| | - Laura Quotti Tubi
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Laboratory, Hematologic Malignancies Unit, Venetian Institute of Molecular Medicine, Padova, Italy
| | - Anna Colpo
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Laboratory, Hematologic Malignancies Unit, Venetian Institute of Molecular Medicine, Padova, Italy
| | - Marco Pizzi
- Department of Medicine, General Pathology and Cytopathology Unit, University of Padova, Padova, Italy
| | - Rocco Cappellesso
- Department of Medicine, General Pathology and Cytopathology Unit, University of Padova, Padova, Italy
| | - Fortunato Zaffino
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Laboratory, Hematologic Malignancies Unit, Venetian Institute of Molecular Medicine, Padova, Italy
| | | | - Anna Cabrelle
- Myeloma and Lymphoma Pathobiology Laboratory, Hematologic Malignancies Unit, Venetian Institute of Molecular Medicine, Padova, Italy
| | - Filippo Marino
- Department of Medicine, General Pathology and Cytopathology Unit, University of Padova, Padova, Italy
| | - Renato Zambello
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Laboratory, Hematologic Malignancies Unit, Venetian Institute of Molecular Medicine, Padova, Italy
| | - Livio Trentin
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Laboratory, Hematologic Malignancies Unit, Venetian Institute of Molecular Medicine, Padova, Italy
| | - Fausto Adami
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy
| | - Carmela Gurrieri
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Laboratory, Hematologic Malignancies Unit, Venetian Institute of Molecular Medicine, Padova, Italy
| | - Gianpietro Semenzato
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Laboratory, Hematologic Malignancies Unit, Venetian Institute of Molecular Medicine, Padova, Italy
- * E-mail: (FP); (GS)
| | - Francesco Piazza
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Laboratory, Hematologic Malignancies Unit, Venetian Institute of Molecular Medicine, Padova, Italy
- * E-mail: (FP); (GS)
| |
Collapse
|
81
|
Shain KH, Tao J. The B-cell receptor orchestrates environment-mediated lymphoma survival and drug resistance in B-cell malignancies. Oncogene 2013; 33:4107-13. [PMID: 24037527 DOI: 10.1038/onc.2013.379] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 07/24/2013] [Accepted: 07/29/2013] [Indexed: 12/14/2022]
Abstract
Specific niches within the lymphoma tumor microenvironment (TME) provide sanctuary for subpopulations of tumor cells through stromal cell-tumor cell interactions. These interactions notably dictate growth, response to therapy and resistance of residual malignant B cells to therapeutic agents. This minimal residual disease (MRD) remains a major challenge in the treatment of B-cell malignancies and contributes to subsequent disease relapse. B-cell receptor (BCR) signaling has emerged as essential mediator of B-cell homing, survival and environment-mediated drug resistance (EMDR). Central to EMDR are chemokine- and integrin-mediated interactions between lymphoma and the TME. Further, stromal cell-B cell adhesion confers a sustained BCR signaling leading to chemokine and integrin activation. Recently, the inhibitors of BCR signaling have garnered a substantial clinical interest because of their effectiveness in B-cell disorders. The efficacy of these agents is, at least in part, attributed to attenuation of BCR-dependent lymphoma-TME interactions. In this review, we discuss the pivotal role of BCR signaling in the integration of intrinsic and extrinsic determinants of TME-mediated lymphoma survival and drug resistance.
Collapse
Affiliation(s)
- K H Shain
- Departments of Malignant Hematology and Chemical Biology and Molecular Medicine Program, H Lee Moffitt Cancer Center and Research Institute at the University of South Florida, Tampa, FL, USA
| | - J Tao
- Departments of Hematopathology and Laboratory Medicine, and Chemical Biology and Molecular Medicine Program, H Lee Moffitt Cancer Center and Research Institute at the University of South Florida, Tampa, FL, USA
| |
Collapse
|
82
|
Hirsch E, Ciraolo E, Franco I, Ghigo A, Martini M. PI3K in cancer-stroma interactions: bad in seed and ugly in soil. Oncogene 2013; 33:3083-90. [PMID: 23893246 DOI: 10.1038/onc.2013.265] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 05/21/2013] [Accepted: 05/21/2013] [Indexed: 02/07/2023]
Abstract
Over the past decade the phosphoinositide-3 kinase (PI3K) signaling pathway emerged as an important player for tumor initiation and growth and, currently, PI3K inhibition constitutes a promising therapeutic approach for solid and hematological tumors. Beside its role in tumor cell evolution, PI3K signaling also provides integral functions for noncancerous cells that reside in healthy tissues surrounding the tumor, also referred as tumor microenvironment (TME). This review will address how PI3K signaling participates to the tumorigenic process and discuss the interaction between tumor cells and the surrounding TME, with particular focus on the role of PI3Ks in tumor-associated immune responses, tumor angiogenesis and metastasis formation.
Collapse
Affiliation(s)
- E Hirsch
- Department of Molecular Biotechnology and Health Sciences-Center for Molecular Biotechnology, University of Torino, Torino, Italy
| | - E Ciraolo
- Department of Molecular Biotechnology and Health Sciences-Center for Molecular Biotechnology, University of Torino, Torino, Italy
| | - I Franco
- Department of Molecular Biotechnology and Health Sciences-Center for Molecular Biotechnology, University of Torino, Torino, Italy
| | - A Ghigo
- Department of Molecular Biotechnology and Health Sciences-Center for Molecular Biotechnology, University of Torino, Torino, Italy
| | - M Martini
- Department of Molecular Biotechnology and Health Sciences-Center for Molecular Biotechnology, University of Torino, Torino, Italy
| |
Collapse
|
83
|
Burger R. Impact of interleukin-6 in hematological malignancies. ACTA ACUST UNITED AC 2013; 40:336-43. [PMID: 24273487 DOI: 10.1159/000354194] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 07/08/2013] [Indexed: 11/19/2022]
Abstract
Almost 3 decades have passed since the discovery and cloning of IL-6, and a tremendous amount of work has contributed to the current knowledge of the biological functions of this cytokine, its receptor, and the signaling pathways that are activated. The understanding of the role of IL-6 in human disease has led to the development of novel therapeutic strategies that block the biological functions of IL-6. In clinical studies, IL-6 and IL-6 receptor antibodies have proven efficacy in rheumatoid arthritis, systemic juvenile idiopathic arthritis, and Castleman's disease, conditions that are known to be driven by IL-6. The focus of this overview is the role of IL-6 in the pathophysiology of hematological malignancies.
Collapse
Affiliation(s)
- Renate Burger
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine, University Hospital Schleswig-Holstein and Christian-Albrechts-University of Kiel, Germany
| |
Collapse
|
84
|
Zhang L, Newberry KJ, Wang M. Ibrutinib: a strong candidate for the future of mantle cell lymphoma treatment. Expert Rev Clin Immunol 2013; 9:495-7. [PMID: 23730878 DOI: 10.1586/eci.13.31] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
85
|
Huang J, Das SK, Jha P, Al Zoughbi W, Schauer S, Claudel T, Sexl V, Vesely P, Birner-Gruenberger R, Kratky D, Trauner M, Hoefler G. The PPARα agonist fenofibrate suppresses B-cell lymphoma in mice by modulating lipid metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:1555-65. [PMID: 23628473 PMCID: PMC4331670 DOI: 10.1016/j.bbalip.2013.04.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 03/28/2013] [Accepted: 04/17/2013] [Indexed: 12/31/2022]
Abstract
Obesity is associated with an increased risk for malignant lymphoma development. We used Bcr/Abl transformed B cells to determine the impact of aggressive lymphoma formation on systemic lipid mobilization and turnover. In wild-type mice, tumor size significantly correlated with depletion of white adipose tissues (WAT), resulting in increased serum free fatty acid (FFA) concentrations which promote B-cell proliferation in vitro. Moreover, B-cell tumor development induced hepatic lipid accumulation due to enhanced hepatic fatty acid (FA) uptake and impaired FA oxidation. Serum triglyceride, FFA, phospholipid and cholesterol levels were significantly elevated. Consistently, serum VLDL/LDL-cholesterol and apolipoprotein B levels were drastically increased. These findings suggest that B-cell tumors trigger systemic lipid mobilization from WAT to the liver and increase VLDL/LDL release from the liver to promote tumor growth. Further support for this concept stems from experiments where we used the peroxisome proliferator-activated receptor α (PPARα) agonist and lipid-lowering drug fenofibrate that significantly suppressed tumor growth independent of angiogenesis and inflammation. In addition to WAT depletion, fenofibrate further stimulated FFA uptake by the liver and restored hepatic FA oxidation capacity, thereby accelerating the clearance of lipids released from WAT. Furthermore, fenofibrate blocked hepatic lipid release induced by the tumors. In contrast, lipid utilization in the tumor tissue itself was not increased by fenofibrate which correlates with extremely low expression levels of PPARα in B-cells. Our data show that fenofibrate associated effects on hepatic lipid metabolism and deprivation of serum lipids are capable to suppress B-cell lymphoma growth which may direct novel treatment strategies. This article is part of a Special Issue entitled Lipid Metabolism in Cancer. B-cell lymphoma induced WAT loss and elevated serum FFA. B-cell lymphoma caused increased liver mass and FA uptake, impaired hepatic FA oxidation and enhanced hepatic lipid export. Fenofibrate reduced lymphoma induced elevation of serum FA by increasing hepatic FA uptake and oxidation. Fenofibrate blocks hepatic lipid export as triglyceride-rich VLDL or cholesterol-rich LDL in B-cell lymphoma bearing mice. Fenofibrate suppresses B-cell lymphoma in mice.
Collapse
Affiliation(s)
- Jianfeng Huang
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Suman Kumar Das
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Pooja Jha
- Institute of Pathology, Medical University of Graz, Graz, Austria
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Wael Al Zoughbi
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Silvia Schauer
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Thierry Claudel
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, Department for Biomedical Sciences, Veterinary University of Vienna, Vienna, Austria
| | - Paul Vesely
- The Scripps Research Institute, La Jolla, CA, USA
| | | | - Dagmar Kratky
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Gerald Hoefler
- Institute of Pathology, Medical University of Graz, Graz, Austria
- Corresponding author at: Institute of Pathology, Medical University of Graz, Auenbruggerplatz 25, 8036 Graz, Austria. Tel.: + 43 316 385 83654; fax: + 43 316 384329.
| |
Collapse
|
86
|
Morscio J, Dierickx D, Tousseyn T. Molecular pathogenesis of B-cell posttransplant lymphoproliferative disorder: what do we know so far? Clin Dev Immunol 2013; 2013:150835. [PMID: 23690819 PMCID: PMC3649442 DOI: 10.1155/2013/150835] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 03/10/2013] [Accepted: 03/11/2013] [Indexed: 12/14/2022]
Abstract
Posttransplant lymphoproliferative disorder (PTLD) is a potentially fatal disease that arises in 2%-10% of solid organ and hematopoietic stem cell transplants and is most frequently of B-cell origin. This very heterogeneous disorder ranges from benign lymphoproliferations to malignant lymphomas, and despite the clear association with Epstein-Barr Virus (EBV) infection, its etiology is still obscure. Although a number of risk factors have been identified (EBV serostatus, graft type, and immunosuppressive regimen), it is currently not possible to predict which transplant patient will eventually develop PTLD. Genetic studies have linked translocations (involving C-MYC, IGH, BCL-2), various copy number variations, DNA mutations (PIM1, PAX5, C-MYC, RhoH/TTF), and polymorphisms in both the host (IFN-gamma, IL-10, TGF-beta, HLA) and the EBV genome to B-cell PTLD development. Furthermore, the tumor microenvironment seems to play an important role in the course of disease representing a local niche that can allow antitumor immune responses even in an immunocompromised host. Taken together, B-cell PTLD pathogenesis is very complex due to the interplay of many different (patient-dependent) factors and requires thorough molecular analysis for the development of novel tailored therapies. This review aims at giving a global overview of the currently known parameters that contribute to the development of B-cell PTLD.
Collapse
Affiliation(s)
- J. Morscio
- KU Leuven, Translational Cell and Tissue Research, Leuven, Belgium
| | - D. Dierickx
- UZ Leuven, Department of Hematology, University Hospitals KU Leuven, Leuven, Belgium
| | - T. Tousseyn
- KU Leuven, Translational Cell and Tissue Research, Leuven, Belgium
- UZ Leuven, Department of Pathology, University Hospitals KU Leuven, Leuven, Belgium
| |
Collapse
|
87
|
Derenzini E, Younes A. Targeting the JAK-STAT pathway in lymphoma: a focus on pacritinib. Expert Opin Investig Drugs 2013; 22:775-85. [PMID: 23442043 DOI: 10.1517/13543784.2013.775244] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION The Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway mediates signaling by cytokine, chemokine and growth factor receptors on cell surface to the nucleus. JAK/STAT pathway is aberrantly activated in a variety of lymphomas, with a dual role of promoting cell survival/proliferation and immune evasion. AREAS COVERED This review describes the preclinical rationale behind the development of JAK inhibitors in lymphoma, some of which are being evaluated in Phase I/II studies, and summarizes the characteristics and clinical results of different JAK inhibitors in clinical development. Available preclinical and clinical data about JAK inhibition in lymphoid malignancies were reviewed using a PubMed access. To date, pacritinib (SB1518), a selective JAK2/FLT3 inhibitor is the first and only JAK inhibitor that has been evaluated in patients with relapsed lymphoma. EXPERT OPINION The preclinical rationale behind the development of pacritinib in lymphoproliferative neoplasms is strong, as the deregulation of the JAK/STAT pathway is involved in the pathogenesis of multiple lymphoma subtypes, although with different mechanisms. Pacritinib demonstrated safety and early clinical efficacy in a variety of lymphoma histologic types, providing the first proof of principle of the potential clinical value of targeting JAK/STAT pathway in lymphoma.
Collapse
Affiliation(s)
- Enrico Derenzini
- University of Bologna, Institute of Hematology and Medical Oncology L.A. Seragnoli, Via Massarenti 9, 40138 Bologna, Italy.
| | | |
Collapse
|