1
|
Chen M, Lan H, Yao S, Jin K, Chen Y. Metabolic Interventions in Tumor Immunity: Focus on Dual Pathway Inhibitors. Cancers (Basel) 2023; 15:cancers15072043. [PMID: 37046703 PMCID: PMC10093048 DOI: 10.3390/cancers15072043] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/23/2023] [Accepted: 03/27/2023] [Indexed: 04/14/2023] Open
Abstract
The metabolism of tumors and immune cells in the tumor microenvironment (TME) can affect the fate of cancer and immune responses. Metabolic reprogramming can occur following the activation of metabolic-related signaling pathways, such as phosphoinositide 3-kinases (PI3Ks) and the mammalian target of rapamycin (mTOR). Moreover, various tumor-derived immunosuppressive metabolites following metabolic reprogramming also affect antitumor immune responses. Evidence shows that intervention in the metabolic pathways of tumors or immune cells can be an attractive and novel treatment option for cancer. For instance, administrating inhibitors of various signaling pathways, such as phosphoinositide 3-kinases (PI3Ks), can improve T cell-mediated antitumor immune responses. However, dual pathway inhibitors can significantly suppress tumor growth more than they inhibit each pathway separately. This review discusses the latest metabolic interventions by dual pathway inhibitors as well as the advantages and disadvantages of this therapeutic approach.
Collapse
Affiliation(s)
- Min Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Huanrong Lan
- Department of Surgical Oncology, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou 310002, China
| | - Shiya Yao
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, China
| | - Ketao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, China
| | - Yun Chen
- Department of Colorectal Surgery, Xinchang People's Hospital, Affiliated Xinchang Hospital, Wenzhou Medical University, Xinchang 312500, China
| |
Collapse
|
2
|
Li Q, Li Z, Luo T, Shi H. Targeting the PI3K/AKT/mTOR and RAF/MEK/ERK pathways for cancer therapy. MOLECULAR BIOMEDICINE 2022; 3:47. [PMID: 36539659 PMCID: PMC9768098 DOI: 10.1186/s43556-022-00110-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 11/10/2022] [Indexed: 12/24/2022] Open
Abstract
The PI3K/AKT/mTOR and RAF/MEK/ERK pathways are commonly activated by mutations and chromosomal translocation in vital targets. The PI3K/AKT/mTOR signaling pathway is dysregulated in nearly all kinds of neoplasms, with the component in this pathway alternations. RAF/MEK/ERK signaling cascades are used to conduct signaling from the cell surface to the nucleus to mediate gene expression, cell cycle processes and apoptosis. RAS, B-Raf, PI3K, and PTEN are frequent upstream alternative sites. These mutations resulted in activated cell growth and downregulated cell apoptosis. The two pathways interact with each other to participate in tumorigenesis. PTEN alterations suppress RAF/MEK/ERK pathway activity via AKT phosphorylation and RAS inhibition. Several inhibitors targeting major components of these two pathways have been supported by the FDA. Dozens of agents in these two pathways have attracted great attention and have been assessed in clinical trials. The combination of small molecular inhibitors with traditional regimens has also been explored. Furthermore, dual inhibitors provide new insight into antitumor activity. This review will further comprehensively describe the genetic alterations in normal patients and tumor patients and discuss the role of targeted inhibitors in malignant neoplasm therapy. We hope this review will promote a comprehensive understanding of the role of the PI3K/AKT/mTOR and RAF/MEK/ERK signaling pathways in facilitating tumors and will help direct drug selection for tumor therapy.
Collapse
Affiliation(s)
- Qingfang Li
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, Chengdu, China
| | - Zhihui Li
- Department of Oncology, The General Hospital of Western Theater Command, Chengdu, PR China
| | - Ting Luo
- Department of Breast, Cancer Center, West China Hospital, Sichuan University, 610041, Chengdu, P. R. China.
| | - Huashan Shi
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, 610041, Chengdu, P. R. China.
| |
Collapse
|
3
|
D’Aloia A, Arrigoni E, Costa B, Berruti G, Martegani E, Sacco E, Ceriani M. RalGPS2 Interacts with Akt and PDK1 Promoting Tunneling Nanotubes Formation in Bladder Cancer and Kidney Cells Microenvironment. Cancers (Basel) 2021; 13:cancers13246330. [PMID: 34944949 PMCID: PMC8699646 DOI: 10.3390/cancers13246330] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/01/2021] [Accepted: 12/14/2021] [Indexed: 12/30/2022] Open
Abstract
Simple Summary Cell-to-cell communication in the tumor microenvironment is a crucial process to orchestrate the different components of the tumoral infrastructure. Among the mechanisms of cellular interplay in cancer cells, tunneling nanotubes (TNTs) are dynamic connections that play an important role. The mechanism of the formation of TNTs among cells and the molecules involved in the process remain to be elucidated. In this study, we analyze several bladder cancer cell lines, representative of tumors at different stages and grades. We demonstrate that TNTs are formed only by mid or high-stage cell lines that show muscle-invasive properties and that they actively transport mitochondria and proteins. The formation of TNTs is triggered by stressful conditions and starts with the assembly of a specific multimolecular complex. In this study, we characterize some of the protein components of the TNTs complex, as they are potential novel molecular targets for future therapies aimed at counteracting tumor progression. Abstract RalGPS2 is a Ras-independent Guanine Nucleotide Exchange Factor for RalA GTPase that is involved in several cellular processes, including cytoskeletal organization. Previously, we demonstrated that RalGPS2 also plays a role in the formation of tunneling nanotubes (TNTs) in bladder cancer 5637 cells. In particular, TNTs are a novel mechanism of cell–cell communication in the tumor microenvironment, playing a central role in cancer progression and metastasis formation. However, the molecular mechanisms involved in TNTs formation still need to be fully elucidated. Here we demonstrate that mid and high-stage bladder cancer cell lines have functional TNTs, which can transfer mitochondria. Moreover, using confocal fluorescence time-lapse microscopy, we show in 5637 cells that TNTs mediate the trafficking of RalA protein and transmembrane MHC class III protein leukocyte-specific transcript 1 (LST1). Furthermore, we show that RalGPS2 is essential for nanotubes generation, and stress conditions boost its expression both in 5637 and HEK293 cell lines. Finally, we prove that RalGPS2 interacts with Akt and PDK1, in addition to LST1 and RalA, leading to the formation of a complex that promotes nanotubes formation. In conclusion, our findings suggest that in the tumor microenvironment, RalGPS2 orchestrates the assembly of multimolecular complexes that drive the formation of TNTs.
Collapse
Affiliation(s)
- Alessia D’Aloia
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (A.D.); (E.A.); (B.C.); (E.M.); (E.S.)
| | - Edoardo Arrigoni
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (A.D.); (E.A.); (B.C.); (E.M.); (E.S.)
| | - Barbara Costa
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (A.D.); (E.A.); (B.C.); (E.M.); (E.S.)
| | - Giovanna Berruti
- Department of Biosciences, University of Milan, Via Celoria 26, 20133 Milan, Italy;
| | - Enzo Martegani
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (A.D.); (E.A.); (B.C.); (E.M.); (E.S.)
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, 20126 Milan, Italy
| | - Elena Sacco
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (A.D.); (E.A.); (B.C.); (E.M.); (E.S.)
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, 20126 Milan, Italy
| | - Michela Ceriani
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (A.D.); (E.A.); (B.C.); (E.M.); (E.S.)
- Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Piazza dell’Ateneo Nuovo 1, 20126 Milano, Italy
- Correspondence: ; Tel.: +39-0264483544
| |
Collapse
|
4
|
Al Dow M, Silveira MAD, Poliquin A, Tribouillard L, Fournier É, Trébaol E, Secco B, Villot R, Tremblay F, Bilodeau S, Laplante M. Control of adipogenic commitment by a STAT3-VSTM2A axis. Am J Physiol Endocrinol Metab 2021; 320:E259-E269. [PMID: 33196296 PMCID: PMC8260376 DOI: 10.1152/ajpendo.00314.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
White adipose tissue (WAT) is a dynamic organ that plays crucial roles in controlling metabolic homeostasis. During development and periods of energy excess, adipose progenitors are recruited and differentiate into adipocytes to promote lipid storage capability. The identity of adipose progenitors and the signals that promote their recruitment are still incompletely characterized. We have recently identified V-set and transmembrane domain-containing protein 2A (VSTM2A) as a novel protein enriched in preadipocytes that amplifies adipogenic commitment. Despite the emerging role of VSTM2A in promoting adipogenesis, the molecular mechanisms regulating Vstm2a expression in preadipocytes are still unknown. To define the molecular mechanisms controlling Vstm2a expression, we have treated preadipocytes with an array of compounds capable of modulating established regulators of adipogenesis. Here, we report that Vstm2a expression is positively regulated by PI3K/mTOR and cAMP-dependent signaling pathways and repressed by the MAPK pathway and the glucocorticoid receptor. By integrating the impact of all the molecules tested, we identified signal transducer and activator of transcription 3 (STAT3) as a novel downstream transcription factor affecting Vstm2a expression. We show that activation of STAT3 increased Vstm2a expression, whereas its inhibition repressed this process. In mice, we found that STAT3 phosphorylation is elevated in the early phases of WAT development, an effect that strongly associates with Vstm2a expression. Our findings identify STAT3 as a key transcription factor regulating Vstm2a expression in preadipocytes.NEW & NOTEWORTHY cAMP-dependent and PI3K-mTOR signaling pathways promote the expression of Vstm2a. STAT3 is a key transcription factor that controls Vstm2a expression in preadipocytes. STAT3 is activated in the early phases of WAT development, an effect that strongly associates with Vstm2a expression.
Collapse
Affiliation(s)
- Manal Al Dow
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, Canada
- Centre de recherche sur le cancer de l'Université Laval, Université Laval, Québec, Canada
| | - Maruhen Amir Datsch Silveira
- Centre de recherche sur le cancer de l'Université Laval, Université Laval, Québec, Canada
- Centre de recherche du CHU de Québec - Université Laval, Québec, Canada
| | - Audrée Poliquin
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, Canada
- Centre de recherche sur le cancer de l'Université Laval, Université Laval, Québec, Canada
| | - Laura Tribouillard
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, Canada
- Centre de recherche sur le cancer de l'Université Laval, Université Laval, Québec, Canada
| | - Éric Fournier
- Centre de recherche sur le cancer de l'Université Laval, Université Laval, Québec, Canada
- Centre de recherche du CHU de Québec - Université Laval, Québec, Canada
- Centre de recherche en données massives de l'Université Laval, Québec, Canada
| | - Eva Trébaol
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, Canada
- Centre de recherche sur le cancer de l'Université Laval, Université Laval, Québec, Canada
| | - Blandine Secco
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, Canada
- Centre de recherche sur le cancer de l'Université Laval, Université Laval, Québec, Canada
- Centre de recherche du CHU de Québec - Université Laval, Québec, Canada
| | - Romain Villot
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, Canada
- Centre de recherche sur le cancer de l'Université Laval, Université Laval, Québec, Canada
| | - Félix Tremblay
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, Canada
- Centre de recherche sur le cancer de l'Université Laval, Université Laval, Québec, Canada
| | - Steve Bilodeau
- Centre de recherche sur le cancer de l'Université Laval, Université Laval, Québec, Canada
- Centre de recherche du CHU de Québec - Université Laval, Québec, Canada
- Centre de recherche en données massives de l'Université Laval, Québec, Canada
- Département de biologie moléculaire, biochimie médicale et pathologie, Faculté de Médecine, Université Laval, Québec, Canada
| | - Mathieu Laplante
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, Canada
- Centre de recherche sur le cancer de l'Université Laval, Université Laval, Québec, Canada
- Département de Médecine, Faculté de Médecine, Université Laval, Québec, Canada
| |
Collapse
|
5
|
Machado S, Silva A, De Sousa-Coelho AL, Duarte I, Grenho I, Santos B, Mayoral-Varo V, Megias D, Sánchez-Cabo F, Dopazo A, Ferreira BI, Link W. Harmine and Piperlongumine Revert TRIB2-Mediated Drug Resistance. Cancers (Basel) 2020; 12:cancers12123689. [PMID: 33316942 PMCID: PMC7763856 DOI: 10.3390/cancers12123689] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 12/02/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Poor survival and treatment failure of patients with cancer are mainly due to resistance to therapy. Tribbles homologue 2 (TRIB2) has recently been identified as a protein that promotes resistance to several anti-cancer drugs. In this study, RNA sequencing and bioinformatics analysis were used with the aim of characterizing the impact of TRIB2 on the expression of genes and developing pharmacological strategies to revert these TRIB2-mediated changes, thereby overcoming therapy resistance. We show that two naturally occurring alkaloids, harmine and piperlongumine, inverse the gene expression profile produced by TRIB2 and sensitize cancer cells to anti-cancer drugs. Our data suggest that harmine and piperlongumine or similar compounds might have the potential to overcome TRIB2-mediated therapy resistance in cancer patients. Abstract Therapy resistance is responsible for most relapses in patients with cancer and is the major challenge to improving the clinical outcome. The pseudokinase Tribbles homologue 2 (TRIB2) has been characterized as an important driver of resistance to several anti-cancer drugs, including the dual ATP-competitive PI3K and mTOR inhibitor dactolisib (BEZ235). TRIB2 promotes AKT activity, leading to the inactivation of FOXO transcription factors, which are known to mediate the cell response to antitumor drugs. To characterize the downstream events of TRIB2 activity, we analyzed the gene expression profiles of isogenic cell lines with different TRIB2 statuses by RNA sequencing. Using a connectivity map-based computational approach, we identified drug-induced gene-expression profiles that invert the TRIB2-associated expression profile. In particular, the natural alkaloids harmine and piperlongumine not only produced inverse gene expression profiles but also synergistically increased BEZ235-induced cell toxicity. Importantly, both agents promote FOXO nuclear translocation without interfering with the nuclear export machinery and induce the transcription of FOXO target genes. Our results highlight the great potential of this approach for drug repurposing and suggest that harmine and piperlongumine or similar compounds might be useful in the clinic to overcome TRIB2-mediated therapy resistance in cancer patients.
Collapse
Affiliation(s)
- Susana Machado
- Centre for Biomedical Research (CBMR), Universidade do Algarve, Campus of Gambelas, Building 8, Room 1.12, 8005-139 Faro, Portugal; (S.M.); (A.S.); (A.L.D.S.-C.); (I.D.); (I.G.); (B.S.)
- Algarve Biomedical Center (ABC), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| | - Andreia Silva
- Centre for Biomedical Research (CBMR), Universidade do Algarve, Campus of Gambelas, Building 8, Room 1.12, 8005-139 Faro, Portugal; (S.M.); (A.S.); (A.L.D.S.-C.); (I.D.); (I.G.); (B.S.)
- Algarve Biomedical Center (ABC), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| | - Ana Luísa De Sousa-Coelho
- Centre for Biomedical Research (CBMR), Universidade do Algarve, Campus of Gambelas, Building 8, Room 1.12, 8005-139 Faro, Portugal; (S.M.); (A.S.); (A.L.D.S.-C.); (I.D.); (I.G.); (B.S.)
- Algarve Biomedical Center (ABC), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| | - Isabel Duarte
- Centre for Biomedical Research (CBMR), Universidade do Algarve, Campus of Gambelas, Building 8, Room 1.12, 8005-139 Faro, Portugal; (S.M.); (A.S.); (A.L.D.S.-C.); (I.D.); (I.G.); (B.S.)
- Algarve Biomedical Center (ABC), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| | - Inês Grenho
- Centre for Biomedical Research (CBMR), Universidade do Algarve, Campus of Gambelas, Building 8, Room 1.12, 8005-139 Faro, Portugal; (S.M.); (A.S.); (A.L.D.S.-C.); (I.D.); (I.G.); (B.S.)
- Algarve Biomedical Center (ABC), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| | - Bruno Santos
- Centre for Biomedical Research (CBMR), Universidade do Algarve, Campus of Gambelas, Building 8, Room 1.12, 8005-139 Faro, Portugal; (S.M.); (A.S.); (A.L.D.S.-C.); (I.D.); (I.G.); (B.S.)
- Algarve Biomedical Center (ABC), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| | - Victor Mayoral-Varo
- Instituto de Investigaciones Biomédicas “Alberto Sols” (CSIC-UAM), Arturo Duperier 4, 28029 Madrid, Spain;
| | - Diego Megias
- Confocal Microscopy Unit, Biotechnology Program, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain;
| | - Fátima Sánchez-Cabo
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; (F.S.-C.); (A.D.)
| | - Ana Dopazo
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; (F.S.-C.); (A.D.)
| | - Bibiana I. Ferreira
- Centre for Biomedical Research (CBMR), Universidade do Algarve, Campus of Gambelas, Building 8, Room 1.12, 8005-139 Faro, Portugal; (S.M.); (A.S.); (A.L.D.S.-C.); (I.D.); (I.G.); (B.S.)
- Algarve Biomedical Center (ABC), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
- Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
- Correspondence: (B.I.F.); (W.L.)
| | - Wolfgang Link
- Instituto de Investigaciones Biomédicas “Alberto Sols” (CSIC-UAM), Arturo Duperier 4, 28029 Madrid, Spain;
- Correspondence: (B.I.F.); (W.L.)
| |
Collapse
|
6
|
Silkenstedt E, Arenas F, Colom-Sanmartí B, Xargay-Torrent S, Higashi M, Giró A, Rodriguez V, Fuentes P, Aulitzky WE, van der Kuip H, Beà S, Toribio ML, Campo E, López-Guerra M, Colomer D. Notch1 signaling in NOTCH1-mutated mantle cell lymphoma depends on Delta-Like ligand 4 and is a potential target for specific antibody therapy. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:446. [PMID: 31676012 PMCID: PMC6825347 DOI: 10.1186/s13046-019-1458-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 10/17/2019] [Indexed: 12/12/2022]
Abstract
Background NOTCH1 gene mutations in mantle cell lymphoma (MCL) have been described in about 5–10% of cases and are associated with significantly shorter survival rates. The present study aimed to investigate the biological impact of this mutation in MCL and its potential as a therapeutic target. Methods Activation of Notch1 signaling upon ligand-stimulation and inhibitory effects of the monoclonal anti-Notch1 antibody OMP-52M51 in NOTCH1-mutated and -unmutated MCL cells were assessed by Western Blot and gene expression profiling. Effects of OMP-52M51 treatment on tumor cell migration and tumor angiogenesis were evaluated with chemotaxis and HUVEC tube formation assays. The expression of Delta-like ligand 4 (DLL4) in MCL lymph nodes was analyzed by immunofluorescence staining and confocal microscopy. A MCL mouse model was used to assess the activity of OMP-52M51 in vivo. Results Notch1 expression can be effectively stimulated in NOTCH1-mutated Mino cells by DLL4, whereas in the NOTCH1-unmutated cell line JeKo-1, less effect was observed upon any ligand-stimulation. DLL4 was expressed by histiocytes in both, NOTCH1-mutated and –unmutated MCL lymph nodes. Treatment of NOTCH1-mutated MCL cells with the monoclonal anti-Notch1 antibody OMP-52M51 effectively prevented DLL4-dependent activation of Notch1 and suppressed the induction of numerous direct Notch target genes involved in lymphoid biology, lymphomagenesis and disease progression. Importantly, in lymph nodes from primary MCL cases with NOTCH1/2 mutations, we detected an upregulation of the same gene sets as observed in DLL4-stimulated Mino cells. Furthermore, DLL4 stimulation of NOTCH1-mutated Mino cells enhanced tumor cell migration and angiogenesis, which could be abolished by treatment with OMP-52M51. Importantly, the effects observed were specific for NOTCH1-mutated cells as they did not occur in the NOTCH1-wt cell line JeKo-1. Finally, we confirmed the potential activity of OMP-52M51 to inhibit DLL4-induced Notch1-Signaling in vivo in a xenograft mouse model of MCL. Conclusion DLL4 effectively stimulates Notch1 signaling in NOTCH1-mutated MCL and is expressed by the microenvironment in MCL lymph nodes. Our results indicate that specific inhibition of the Notch1-ligand-receptor interaction might provide a therapeutic alternative for a subset of MCL patients.
Collapse
Affiliation(s)
- Elisabeth Silkenstedt
- Experimental Therapeutics in Lymphoid Malignancies Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Department of Internal Medicine III, University Hospital, Ludwig Maximilian University, Munich, Germany.,Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology and University of Tuebingen, Stuttgart, Germany
| | - Fabian Arenas
- Experimental Therapeutics in Lymphoid Malignancies Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Berta Colom-Sanmartí
- Experimental Therapeutics in Lymphoid Malignancies Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Sílvia Xargay-Torrent
- Experimental Therapeutics in Lymphoid Malignancies Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Morihiro Higashi
- Experimental Therapeutics in Lymphoid Malignancies Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Ariadna Giró
- Experimental Therapeutics in Lymphoid Malignancies Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Vanina Rodriguez
- Experimental Therapeutics in Lymphoid Malignancies Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Patricia Fuentes
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - Walter E Aulitzky
- Department of Hematology and Oncology, Robert-Bosch-Hospital, Stuttgart, Germany
| | - Heiko van der Kuip
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology and University of Tuebingen, Stuttgart, Germany
| | - Sílvia Beà
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Lymphoid Neoplasm Program, IDIBAPS, Barcelona, Spain
| | - Maria L Toribio
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - Elias Campo
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Lymphoid Neoplasm Program, IDIBAPS, Barcelona, Spain.,Hematopathology Section, Hospital Clínic, Barcelona, Spain.,University of Barcelona, Barcelona, Spain
| | - Mònica López-Guerra
- Experimental Therapeutics in Lymphoid Malignancies Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Hematopathology Section, Hospital Clínic, Barcelona, Spain
| | - Dolors Colomer
- Experimental Therapeutics in Lymphoid Malignancies Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain. .,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain. .,Hematopathology Section, Hospital Clínic, Barcelona, Spain. .,University of Barcelona, Barcelona, Spain.
| |
Collapse
|
7
|
Specific NOTCH1 antibody targets DLL4-induced proliferation, migration, and angiogenesis in NOTCH1-mutated CLL cells. Oncogene 2019; 39:1185-1197. [PMID: 31616059 PMCID: PMC7002297 DOI: 10.1038/s41388-019-1053-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 09/10/2019] [Accepted: 10/01/2019] [Indexed: 11/30/2022]
Abstract
Targeting Notch signaling has emerged as a promising therapeutic strategy for chronic lymphocytic leukemia (CLL), particularly in NOTCH1-mutated patients. We provide first evidence that the Notch ligand DLL4 is a potent stimulator of Notch signaling in NOTCH1-mutated CLL cells while increases cell proliferation. Importantly, DLL4 is expressed in histiocytes from the lymph node, both in NOTCH1-mutated and -unmutated cases. We also show that the DLL4-induced activation of the Notch signaling pathway can be efficiently blocked with the specific anti-Notch1 antibody OMP-52M51. Accordingly, OMP-52M51 also reverses Notch-induced MYC, CCND1, and NPM1 gene expression as well as cell proliferation in NOTCH1-mutated CLL cells. In addition, DLL4 stimulation triggers the expression of protumor target genes, such as CXCR4, NRARP, and VEGFA, together with an increase in cell migration and angiogenesis. All these events can be antagonized by OMP-52M51. Collectively, our results emphasize the role of DLL4 stimulation in NOTCH1-mutated CLL and confirm the specific therapeutic targeting of Notch1 as a promising approach for this group of poor prognosis CLL patients.
Collapse
|
8
|
Diamond B, Kumar A. Mantle Cell Lymphoma: Current and Emerging Treatment Strategies and Unanswered Questions. Hematol Oncol Clin North Am 2019; 33:613-626. [PMID: 31229158 DOI: 10.1016/j.hoc.2019.03.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Mantle cell lymphoma (MCL) is biologically and clinically heterogeneous with no clear standard of care. Overexpression of cyclin D1 is a hallmark of MCL. Evolving characterization of other molecular drivers explain a variety of disease phenotypes. These molecular profiles challenge risk stratification techniques. TP53-deleted disease is associated with adverse outcomes. Frontline treatment programs include intensive chemoimmunotherapy and autologous stem cell transplantation. Minimal residual disease may change management of MCL and guide therapy. As commonly dysregulated pathways become enumerated, novel biologically targeted agents and their combinations have been developed that will increasingly replace older, more toxic, and less efficacious regimens.
Collapse
Affiliation(s)
- Benjamin Diamond
- Lymphoma Department, MSKCC, 1275 York Avenue, Box 468, New York, NY 10065, USA; Department of Medicine, Medical Oncology, New York, NY, USA.
| | - Anita Kumar
- Lymphoma Department, MSKCC, 1275 York Avenue, Box 468, New York, NY 10065, USA; Department of Medicine, Lymphoma Service, New York, NY, USA
| |
Collapse
|
9
|
Pham LV, Pogue E, Ford RJ. The Role of Macrophage/B-Cell Interactions in the Pathophysiology of B-Cell Lymphomas. Front Oncol 2018; 8:147. [PMID: 29868471 PMCID: PMC5951963 DOI: 10.3389/fonc.2018.00147] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/23/2018] [Indexed: 12/19/2022] Open
Abstract
Macrophages (MPs) are heterogeneous, multifunctional, myeloid-derived leukocytes that are part of the innate immune system, playing wide-ranging critical roles in basic biological activities, including maintenance of tissue homeostasis involving clearance of microbial pathogens. Tumor-associated MPs (TAMs) are MPs with defined specific M2 phenotypes now known to play central roles in the pathophysiology of a wide spectrum of malignant neoplasms. Also, TAMs are often intrinsic cellular components of the essential tumor microenvironment (TME). In concert with lymphoid-lineage B and T cells at various developmental stages, TAMs can mediate enhanced tumor progression, often leading to poor clinical prognosis, at least partly through secretion of chemokines, cytokines, and various active proteases shown to stimulate tumor growth, angiogenesis, metastasis, and immunosuppression. Researchers recently showed that TAMs express certain key checkpoint-associated proteins [e.g., programmed cell death protein 1 (PD-1), programmed cell death-ligand 1 (PD-L1)] that appear to be involved in T-cell activation and that these proteins are targets of other specific checkpoint-blocking immunotherapies (anti-PD-1/PD-L1) currently part of new therapeutic paradigms for chemotherapy-resistant neoplasms. Although much is known about the wide spectrum and flexibility of MPs under many normal and neoplastic conditions, relatively little is known about the increasingly important interactions between MPs and B-lymphoid cells, particularly in the TME in patients with aggressive B-cell non-Hodgkin lymphoma (NHL-B). Normal and neoplastic lymphoid and myeloid cell/MP lineages appear to share many primitive cellular characteristics as well as transcriptional factor interactions in human and animal ontogenic studies. Such cells are capable of ectopic transcription factor-induced lineage reprogramming or transdifferentiation from early myeloid/monocytic lineages to later induce B-cell lymphomagenesis in experimental in vivo murine systems. Close cellular interactions between endogenous clonal neoplastic B cells and related aberrant myeloid precursor cells/MPs appear to be important interactive components of aggressive NHL-B that we discuss herein in the larger context of the putative role of B-cell/MP cellular lineage interactions involved in NHL-B pathophysiology during ensuing lymphoma development.
Collapse
Affiliation(s)
- Lan V Pham
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Elizabeth Pogue
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Richard J Ford
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
10
|
Inamdar AA, Goy A, Ayoub NM, Attia C, Oton L, Taruvai V, Costales M, Lin YT, Pecora A, Suh KS. Mantle cell lymphoma in the era of precision medicine-diagnosis, biomarkers and therapeutic agents. Oncotarget 2018; 7:48692-48731. [PMID: 27119356 PMCID: PMC5217048 DOI: 10.18632/oncotarget.8961] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 04/10/2016] [Indexed: 12/15/2022] Open
Abstract
Despite advances in the development of clinical agents for treating Mantle Cell Lymphoma (MCL), treatment of MCL remains a challenge due to complexity and frequent relapse associated with MCL. The incorporation of conventional and novel diagnostic approaches such as genomic sequencing have helped improve understanding of the pathogenesis of MCL, and have led to development of specific agents targeting signaling pathways that have recently been shown to be involved in MCL. In this review, we first provide a general overview of MCL and then discuss about the role of biomarkers in the pathogenesis, diagnosis, prognosis, and treatment for MCL. We attempt to discuss major biomarkers for MCL and highlight published and ongoing clinical trials in an effort to evaluate the dominant signaling pathways as drugable targets for treating MCL so as to determine the potential combination of drugs for both untreated and relapse/refractory cases. Our analysis indicates that incorporation of biomarkers is crucial for patient stratification and improve diagnosis and predictability of disease outcome thus help us in designing future precision therapies. The evidence indicates that a combination of conventional chemotherapeutic agents and novel drugs designed to target specific dysregulated signaling pathways can provide the effective therapeutic options for both untreated and relapse/refractory MCL.
Collapse
Affiliation(s)
- Arati A Inamdar
- The Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Andre Goy
- Clinical Divisions, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Nehad M Ayoub
- Department of Clinical Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Christen Attia
- The Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Lucia Oton
- The Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Varun Taruvai
- The Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Mark Costales
- The Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Yu-Ting Lin
- The Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Andrew Pecora
- Clinical Divisions, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - K Stephen Suh
- The Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| |
Collapse
|
11
|
Wu D, Cheng J, Sun G, Wu S, Li M, Gao Z, Zhai S, Li P, Su D, Wang X. p70S6K promotes IL-6-induced epithelial-mesenchymal transition and metastasis of head and neck squamous cell carcinoma. Oncotarget 2017; 7:36539-36550. [PMID: 27174914 PMCID: PMC5095019 DOI: 10.18632/oncotarget.9282] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 04/24/2016] [Indexed: 12/14/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the fifth most common cancer worldwide and a common cause of cancer-related death, with a 5-year survival rate of less than 60%. IL-6 has been suggested to play an important role in cancer metastasis, but its mechanism in HNSCC has not been fully clarified. p70S6K has been reported to induce epithelial-mesenchymal transition (EMT) of ovarian cancer, but its role in HNSCC remains unknown. In this study, we found that p70S6K and IL-6 were upregulated in high-metastatic HNSCC cell lines that underwent EMT when compared to paired low-metastatic cell lines. Overexpression of p70S6K promoted EMT and migration of HNSCC cells, while downregulation of p70S6K attenuated IL-6-induced EMT and cell migration. Furthermore, IL-6-induced p70S6K activation was attenuated by inhibitors of the PI3K/Akt/mTOR, MAPK/ERK, and JAK/STAT3 signaling pathways, suggesting that it located downstream of these pathways. These findings suggest that p70S6K promotes IL-6-induced EMT and metastasis of HNSCC. Targeting p70S6K for HNSCC therapy may benefit patients through the inhibition of tumor growth, as well as metastasis.
Collapse
Affiliation(s)
- Dandan Wu
- Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China.,Department of Basic Medicine, Kangda College of Nanjing Medical University, Lianyungang, Jiangsu Province 222000, China
| | - Jie Cheng
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Geng Sun
- Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Shengjie Wu
- Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Min Li
- Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Zhongyuan Gao
- Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Sulan Zhai
- Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Ping Li
- Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Dongming Su
- Center for Clinical Pathology and Laboratory, Affiliated Hospital of Yifu, Nanjing Medical University, Nanjing, Jiangsu Province 211166, China.,Department of Pathology, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Xuerong Wang
- Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China.,Center for Clinical Pathology and Laboratory, Affiliated Hospital of Yifu, Nanjing Medical University, Nanjing, Jiangsu Province 211166, China.,Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| |
Collapse
|
12
|
Huang X, Cao M, Wu S, Wang L, Hu J, Mehran RJ, Roth JA, Swisher SG, Wang RY, Kantarjian HM, Andreeff M, Sun X, Fang B. Anti-leukemia activity of NSC-743380 in SULT1A1-expressing acute myeloid leukemia cells is associated with inhibitions of cFLIP expression and PI3K/AKT/mTOR activities. Oncotarget 2017; 8:102150-102160. [PMID: 29254232 PMCID: PMC5731942 DOI: 10.18632/oncotarget.22235] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 09/30/2017] [Indexed: 11/25/2022] Open
Abstract
Our recent study showed that acute myeloid leukemia (AML) cells expressing SULT1A1 are highly sensitive to NSC-743380, a small molecule that inhibits STAT3 activity and induces SULT1A1-dependent apoptosis of various cancer cell lines. In this study, we characterized the molecular mechanisms of NSC-743380-mediated anti-leukemia activity in AML cell lines and antileukemia activity of NSC-743380 in patient-derived primary leukemia cells from AML patients. Our results showed that treatment with NSC-743380 triggered robust apoptosis in SULT1A1-positive AML cells. Treatment with NSC-743380 did not increase intracellular reactive oxygen species or change of STAT3 activity in AML cells, but did dramatically and rapidly decrease cFLIP expression. Proteomic analysis with reverse phase protein microarray revealed that treatment of U937 and THP-1 AML cells with NSC-743380 led to drastic and time-dependent suppression of phosphorylation of several key nodes in the PI3K/AKT/mTOR pathway, including AKT and mTOR. Moreover, primary AML cells expressed SULT1A1 were highly sensitive to treatment with NSC-743380, which was not affected by co-culture with bone marrow mesenchymal stem cells. Thus, our results provide proof-of-concept evidence that AML cells expressing SULT1A1 can be targeted by small molecules that induce apoptosis through inhibiting the expression or activities of multiple targets.
Collapse
Affiliation(s)
- Xiao Huang
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Mengru Cao
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Shuhong Wu
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Li Wang
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Jing Hu
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Reza J. Mehran
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Jack A. Roth
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Stephen G. Swisher
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Rui-Yu Wang
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Hagop M. Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Michael Andreeff
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Xiaoping Sun
- Department of Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Bingliang Fang
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
13
|
SOX11 promotes tumor protective microenvironment interactions through CXCR4 and FAK regulation in mantle cell lymphoma. Blood 2017; 130:501-513. [PMID: 28533307 DOI: 10.1182/blood-2017-04-776740] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 05/16/2017] [Indexed: 12/15/2022] Open
Abstract
SOX11 overexpression in mantle cell lymphoma (MCL) has been associated with more aggressive behavior and worse outcome. However, SOX11 oncogenic pathways driving MCL tumor progression are poorly understood. Here, we demonstrate that SOX11 binds to regulatory regions of 2 important genes for microenvironment signals in cancer: (C-X-C motif) chemokine receptor 4 (CXCR4) and PTK2 (encoding for focal adhesion kinase [FAK]). Moreover, SOX11+ xenograft and human primary MCL tumors overexpress cell migration and stromal stimulation gene signatures compared with their SOX11- counterparts. We show that SOX11 directly upregulates CXCR4 and FAK expression, activating PI3K/AKT and ERK1/2 FAK-downstream pathways in MCL. Concordantly, SOX11+ MCL cells have higher cell migration, transmigration through endothelial cells, adhesion to stromal cells, and cell proliferation and display an increased resistance to conventional drug therapies compared with SOX11- MCL cells. Specific FAK inhibition blocks downstream PI3K/AKT- and ERK1/2-mediated phosphorylation. Additionally, specific FAK and PI3K inhibitors reduce SOX11-enhanced MCL cell migration and stromal interactions and revert cell adhesion-mediated drug resistance (CAM-DR) to the same levels as SOX11- MCL cells. In intravenous MCL xenograft models, SOX11+ MCL cells display higher cell migration, invasion, and growth compared with SOX11-knockdown cells, and specific FAK and CXCR4 inhibitors impair SOX11-enhanced MCL engraftment in bone marrow. Overall, our results suggest that SOX11 promotes MCL homing and invasion and increases CAM-DR through the direct regulation of CXCR4 and FAK expression and FAK/PI3K/AKT pathway activation, contributing to a more aggressive phenotype. Inhibition of this pathway may represent an efficient strategy to overcome stromal-mediated chemotherapy refractoriness in aggressive MCL.
Collapse
|
14
|
Enhanced Anticancer Activity of PF-04691502, a Dual PI3K/mTOR Inhibitor, in Combination With VEGF siRNA Against Non-small-cell Lung Cancer. MOLECULAR THERAPY-NUCLEIC ACIDS 2016; 5:e384. [PMID: 27845769 PMCID: PMC5155322 DOI: 10.1038/mtna.2016.90] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 09/08/2016] [Indexed: 12/29/2022]
Abstract
Lung cancer is the leading cause of cancer deaths in both men and women in the United States accounting for about 27% of all cancer deceases. In our effort to develop newer therapy for lung cancer, we evaluated the combinatory antitumor effect of siRNA targeting VEGF and the PI3K/mTOR dual inhibitor PF-04691502. We analyzed the anticancer effect of siRNA VEGF and PF-04691502 combination on proliferation, colony formation and migration of A549 and H460 lung cancer cells. Additionally, we assessed the combination treatment antiangiogenic effect on human umbilical vein endothelial cells. Here, we show for the first time that the antiangiogenic siRNA VEGF potentiates the PF-04691502 anticancer activity against non–small-cell lung cancer. We observed a significant (P < 0.05) decrease in cell viability, colony formation, and migration for the combination comparing with the single drug treatment. We also showed a significant (P < 0.05) enhanced effect of the combination treatment inhibiting angiogenesis progression and tube formation organization compared to the single drug treatment groups. Our findings demonstrated an enhanced synergistic anticancer effect of siRNA VEGF and PF-04691502 combination therapy by targeting two main pathways involved in lung cancer cell survival and angiogenesis which will be useful for future preclinical studies and potentially for lung cancer patient management.
Collapse
|
15
|
Montraveta A, Xargay-Torrent S, Rosich L, López-Guerra M, Roldán J, Rodríguez V, Lee-Vergés E, de Frías M, Campàs C, Campo E, Roué G, Colomer D. Bcl-2high mantle cell lymphoma cells are sensitized to acadesine with ABT-199. Oncotarget 2016; 6:21159-72. [PMID: 26110568 PMCID: PMC4673257 DOI: 10.18632/oncotarget.4230] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 05/13/2015] [Indexed: 11/25/2022] Open
Abstract
Acadesine is a nucleoside analogue with known activity against B-cell malignancies. Herein, we showed that in mantle cell lymphoma (MCL) cells acadesine induced caspase-dependent apoptosis through turning on the mitochondrial apoptotic machinery. At the molecular level, the compound triggered the activation of the AMPK pathway, consequently modulating known downstream targets, such as mTOR and the cell motility-related vasodilator-stimulated phosphoprotein (VASP). VASP phosphorylation by acadesine was concomitant with a blockade of CXCL12-induced migration. The inhibition of the mTOR cascade by acadesine, committed MCL cells to enter in apoptosis by a translational downregulation of the antiapoptotic Mcl-1 protein. In contrast, Bcl-2 protein levels were unaffected by acadesine and MCL samples expressing high levels of Bcl-2 tended to have a reduced response to the drug. Targeting Bcl-2 with the selective BH3-mimetic agent ABT-199 sensitized Bcl-2 high MCL cells to acadesine. This effect was validated in vivo, where the combination of both agents displayed a more marked inhibition of tumor outgrowth than each drug alone. These findings support the notions that antiapoptotic proteins of the Bcl-2 family regulate MCL cell sensitivity to acadesine and that the combination of this agent with Bcl-2 inhibitors might be an interesting therapeutic option to treat MCL patients.
Collapse
Affiliation(s)
- Arnau Montraveta
- Experimental Therapeutics in Lymphoid Malignancies Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Sílvia Xargay-Torrent
- Experimental Therapeutics in Lymphoid Malignancies Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Laia Rosich
- Experimental Therapeutics in Lymphoid Malignancies Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Mònica López-Guerra
- Experimental Therapeutics in Lymphoid Malignancies Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Unitat d'Hematopatologia, Hospital Clinic, IDIBAPS, Barcelona, Spain
| | - Jocabed Roldán
- Experimental Therapeutics in Lymphoid Malignancies Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Vanina Rodríguez
- Experimental Therapeutics in Lymphoid Malignancies Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Eriong Lee-Vergés
- Experimental Therapeutics in Lymphoid Malignancies Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Mercè de Frías
- Advancell-Advanced In Vitro Cell Technologies S.A., Barcelona, Spain
| | - Clara Campàs
- Advancell-Advanced In Vitro Cell Technologies S.A., Barcelona, Spain
| | - Elias Campo
- Unitat d'Hematopatologia, Hospital Clinic, IDIBAPS, Barcelona, Spain
| | - Gaël Roué
- Experimental Therapeutics in Lymphoid Malignancies Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Dolors Colomer
- Experimental Therapeutics in Lymphoid Malignancies Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Unitat d'Hematopatologia, Hospital Clinic, IDIBAPS, Barcelona, Spain
| |
Collapse
|
16
|
Awan FT, Gore L, Gao L, Sharma J, Lager J, Costa LJ. Phase Ib trial of the PI3K/mTOR inhibitor voxtalisib (SAR245409) in combination with chemoimmunotherapy in patients with relapsed or refractory B-cell malignancies. Br J Haematol 2016; 175:55-65. [PMID: 27293194 DOI: 10.1111/bjh.14181] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 04/15/2016] [Indexed: 01/04/2023]
Abstract
This phase Ib, dose-escalation study investigated the maximum tolerated dose (MTD), recommended phase II dose (RP2D), safety, pharmacokinetics (PK) and preliminary efficacy of the pan-class I phosphoinositide 3-kinase (PI3K) and mechanistic target of rapamycin (mTOR) inhibitor voxtalisib [30 or 50 mg twice daily (BID)], in combination with rituximab (voxtalisib+rituximab) or rituximab plus bendamustine (voxtalisib+rituximab+bendamustine), in relapsed or refractory indolent B-cell non-Hodgkin lymphoma (NHL), mantle cell lymphoma and chronic lymphocytic leukaemia (CLL). MTD and RP2D of voxtalisib were determined using a 3 + 3 dose-escalation design. Adverse events (AEs), plasma PK and disease response were recorded. Thirty-seven patients were enrolled. The RP2D of voxtalisib in combination with rituximab or rituximab+bendamustine was 50 mg BID. Four patients experienced a total of five dose-limiting toxicities. The most frequent AEs were nausea (45·9%), fatigue (37·8%) headache (32·4%) and pyrexia (32·4%). The most frequent grade ≥3 AEs were neutropenia (27·0%), thrombocytopenia (24·3%), anaemia (16·2%) and febrile neutropenia (10·8%). Voxtalisib PK parameters were not affected by co-administration with rituximab or rituximab+bendamustine. Of 35 efficacy-evaluable patients, four (11·4%) achieved complete response and 13 (37·1%) achieved partial response. Voxtalisib, in combination with rituximab or rituximab+bendamustine, demonstrated an acceptable safety profile and encouraging anti-tumour activity in relapsed or refractory B-cell malignancies.
Collapse
Affiliation(s)
- Farrukh T Awan
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.
| | - Lia Gore
- Developmental Therapeutics Program, University of Colorado Comprehensive Cancer Center, Aurora, CO, USA
| | | | | | | | | |
Collapse
|
17
|
Akhenblit PJ, Hanke NT, Gill A, Persky DO, Howison CM, Pagel MD, Baker AF. Assessing Metabolic Changes in Response to mTOR Inhibition in a Mantle Cell Lymphoma Xenograft Model Using AcidoCEST MRI. Mol Imaging 2016; 15:15/0/1536012116645439. [PMID: 27140422 PMCID: PMC4878391 DOI: 10.1177/1536012116645439] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 02/23/2016] [Indexed: 01/16/2023] Open
Abstract
AcidoCEST magnetic resonance imaging (MRI) has previously been shown to measure tumor extracellular pH (pHe) with excellent accuracy and precision. This study investigated the ability of acidoCEST MRI to monitor changes in tumor pHe in response to therapy. To perform this study, we used the Granta 519 human mantle cell lymphoma cell line, which is an aggressive B-cell malignancy that demonstrates activation of the phosphatidylinositol-3-kinase/Akt/mammalian target of rapamycin (mTOR) pathway. We performed in vitro and in vivo studies using the Granta 519 cell line to investigate the efficacy and associated changes induced by the mTOR inhibitor, everolimus (RAD001). AcidoCEST MRI studies showed a statistically significant increase in tumor pHe of 0.10 pH unit within 1 day of initiating treatment, which foreshadowed a decrease in tumor growth of the Granta 519 xenograft model. AcidoCEST MRI then measured a decrease in tumor pHe 7 days after initiating treatment, which foreshadowed a return to normal tumor growth rate. Therefore, this study is a strong example that acidoCEST MRI can be used to measure tumor pHe that may serve as a marker for therapeutic efficacy of anticancer therapies.
Collapse
Affiliation(s)
- Paul J Akhenblit
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ, USA
| | - Neale T Hanke
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA
| | - Alexander Gill
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA
| | - Daniel O Persky
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA
| | | | - Mark D Pagel
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ, USA University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA Department of Medical Imaging, University of Arizona, Tucson, AZ, USA
| | - Amanda F Baker
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
18
|
Ferreira ACDS, de-Freitas-Junior JCM, Morgado-Díaz JA, Ridley AJ, Klumb CE. Dual inhibition of histone deacetylases and phosphoinositide 3-kinases: effects on Burkitt lymphoma cell growth and migration. J Leukoc Biol 2016; 99:569-78. [PMID: 26561567 DOI: 10.1189/jlb.2a0415-162r] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 10/19/2015] [Indexed: 02/04/2023] Open
Abstract
Burkitt lymphoma is a highly aggressive non-Hodgkin lymphoma that is characterized by MYC deregulation. Recently, the PI3K pathway has emerged as a cooperative prosurvival mechanism in Burkitt lymphoma. Despite the highly successful results of treatment that use high-dose chemotherapy regimens in pediatric Burkitt lymphoma patients, the survival rate of pediatric patients with progressive or recurrent disease is low. PI3Ks are also known to regulate cell migration, and abnormal cell migration may contribute to cancer progression and dissemination in Burkitt lymphoma. Little is known about Burkitt lymphoma cell migration, but the cooperation between MYC and PI3K in Burkitt lymphoma pathogenesis suggests that a drug combination could be used to target the different steps involved in Burkitt lymphoma cell dissemination and disease progression. The aim of this study was to investigate the effects of the histone deacetylase inhibitor suberoylanilide hydroxamic acid combined with the PI3K inhibitor LY294002 on Burkitt lymphoma cell growth and migration. The combination enhanced the cell growth inhibition and cell-cycle arrest induced by the PI3K inhibitor or histone deacetylase inhibitor individually. Moreover, histone deacetylase inhibitor/PI3K inhibitor cotreatment suppressed Burkitt lymphoma cell migration and decreased cell polarization, Akt and ERK1/2 phosphorylation, and leads to RhoB induction. In summary, the histone deacetylase inhibitor/PI3Ki combination inhibits cell proliferation and migration via alterations in PI3K signaling and histone deacetylase activity, which is involved in the acetylation of α-tubulin and the regulation of RhoB expression.
Collapse
Affiliation(s)
- Ana Carolina dos Santos Ferreira
- *Programa de Pesquisa em Hemato-Oncologia Molecular, Laboratório de Hemato-oncologia Celular e Molecular, and Programa de Biologia Celular, Laboratório de Biologia Estrutural-Instituto Nacional de Câncer, Rio de Janeiro, Brazil; and Randall Division of Cell and Molecular Biophysics, King's College London, United Kingdom
| | - Julio Cesar Madureira de-Freitas-Junior
- *Programa de Pesquisa em Hemato-Oncologia Molecular, Laboratório de Hemato-oncologia Celular e Molecular, and Programa de Biologia Celular, Laboratório de Biologia Estrutural-Instituto Nacional de Câncer, Rio de Janeiro, Brazil; and Randall Division of Cell and Molecular Biophysics, King's College London, United Kingdom
| | - Jose Andres Morgado-Díaz
- *Programa de Pesquisa em Hemato-Oncologia Molecular, Laboratório de Hemato-oncologia Celular e Molecular, and Programa de Biologia Celular, Laboratório de Biologia Estrutural-Instituto Nacional de Câncer, Rio de Janeiro, Brazil; and Randall Division of Cell and Molecular Biophysics, King's College London, United Kingdom
| | - Anne J Ridley
- *Programa de Pesquisa em Hemato-Oncologia Molecular, Laboratório de Hemato-oncologia Celular e Molecular, and Programa de Biologia Celular, Laboratório de Biologia Estrutural-Instituto Nacional de Câncer, Rio de Janeiro, Brazil; and Randall Division of Cell and Molecular Biophysics, King's College London, United Kingdom
| | - Claudete Esteves Klumb
- *Programa de Pesquisa em Hemato-Oncologia Molecular, Laboratório de Hemato-oncologia Celular e Molecular, and Programa de Biologia Celular, Laboratório de Biologia Estrutural-Instituto Nacional de Câncer, Rio de Janeiro, Brazil; and Randall Division of Cell and Molecular Biophysics, King's College London, United Kingdom
| |
Collapse
|
19
|
Jin Z, Qing K, Ouyang Y, Liu Z, Wang W, Li X, Xu Z, Li J. Low dose of lenalidmide and PI3K/mTOR inhibitor trigger synergistic cytoxicity in activated B cell-like subtype of diffuse large B cell lymphoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2016; 35:52. [PMID: 27009084 PMCID: PMC4806505 DOI: 10.1186/s13046-016-0327-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 03/17/2016] [Indexed: 12/16/2022]
Abstract
Background Activated B cell-like subtype of diffuse large B cell lymphoma (ABC-DLBCL) presents aggressive clinical courses and poor prognosis. Targeting key pathways may raise the possibility of improving clinical outcomes. Methods The synergetic effects were assessed by CCK-8 assay and measured by isobologram analysis. The NVP-Bez235 and lenalidomide cytotoxicity were measured by flow cytometry, Western Blot and si-RNA transfection. The combined treatment inducing tumor regression in vivo was performed in nude mice of OCI-Ly10 xenograft mouse model. Results Low dose of two agents represented significant inhibition of proliferation with CI value < 1. NVP-Bez235 combined with lenalidomide remarkably increased apoptosis through intrinsic pathway by upregulating Bim, Bax and downregulating Bcl-xL. Akt, especially NF-κB, played an important role in the synergetic effects. Cotreatment also induced the cell cycle to be arrested in G0/G1 phase, and decreased S phase by increasing p21 expression, downregulating cyclinA and diminishing CDK2 phosphorylation in Su-DHL2 and OCI-Ly3 but not in OCI-Ly10. Mice treated with NVP-Bez235/lenalidomide represented obvious tumor growth regression and prolonged overall survival. Conclusions Our findings demonstrated the synergistic effect of low dose of NVP-Bez235 and lenalidomide in ABC-DLBCL, the underlying mechanism may be multifunctional, involving apoptosis, Akt and NF-κB inactivation and cell cycle arrest. Cotreatment was also effective in vivo. These data pave the way for potential treatment of ABC-DLBCL with combination of NVP-Bez235 and lenalidomide.
Collapse
Affiliation(s)
- Zhen Jin
- Shanghai Institute of Hematology, State Key Laboratory for Medical Genomics, Ruijin Hospital affiliated to School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kai Qing
- Shanghai Institute of Hematology, State Key Laboratory for Medical Genomics, Ruijin Hospital affiliated to School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuan Ouyang
- Shanghai Institute of Hematology, State Key Laboratory for Medical Genomics, Ruijin Hospital affiliated to School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhao Liu
- Department of Hematology, Shanghai Institute of Hematology, Ruijin Hospital affiliated to School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wenfang Wang
- Shanghai Institute of Hematology, State Key Laboratory for Medical Genomics, Ruijin Hospital affiliated to School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoyang Li
- Department of Hematology, Shanghai Institute of Hematology, Ruijin Hospital affiliated to School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zizhen Xu
- Department of Laboratory Medicine, Ruijin Hospital affiliated to School of Medicine, Shanghai Jiao Tong University, 197 Rui Jin Er Road, Shanghai, China.
| | - Junmin Li
- Department of Hematology, Shanghai Institute of Hematology, Ruijin Hospital affiliated to School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
20
|
Rauert-Wunderlich H, Rudelius M, Ott G, Rosenwald A. Targeting protein kinase C in mantle cell lymphoma. Br J Haematol 2016; 173:394-403. [PMID: 26914495 DOI: 10.1111/bjh.13973] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 12/21/2015] [Indexed: 12/18/2022]
Abstract
Although targeting the Bruton tyrosine kinase (BTK) with ibrutinib has changed lymphoma treatment, patients with mantle cell lymphoma (MCL) remain incurable. In this study, we characterized a broad range of MCL cell lines and primary MCL cells with respect to the response to the BTK inhibitor, ibrutinib, and compared it with the response to the protein kinase C (PKC) inhibitor, sotrastaurin. At clinically relevant concentrations, each drug induced potent cell death only in the REC-1 cell line, which was accompanied by robust inhibition of AKT and ERK1/ERK2 (ERK1/2, also termed MAPK3/MAPK1) phosphorylation. In sensitive REC-1 cells, the drug-mediated impaired phosphorylation was obvious on the levels of B-cell receptor-induced and basal phosphorylation. Similar results were obtained in primary MCL cells with ibrutinib and in a subset with sotrastaurin. The various drug-resistant MCL cell lines showed very distinct responses in terms of basal AKT and ERK1/2 phosphorylation. Interestingly, targeting PKC and BTK at the same time led to ibrutinib-mediated rescue of a weak sotrastaurin-induced apoptosis in MINO cells. Additional targeting of AKT sensitized MINO cells to inhibitor-mediated cytotoxicity. In summary, MCL cells are heterogeneous in their response to BTK or PKC inhibition, indicating the need for even more individualized targeted treatment approaches in subsets of MCL patients.
Collapse
Affiliation(s)
- Hilka Rauert-Wunderlich
- Institute of Pathology, University of Würzburg and Comprehensive Cancer Center (CCC) Mainfranken, Würzburg, Germany
| | - Martina Rudelius
- Institute of Pathology, University of Würzburg and Comprehensive Cancer Center (CCC) Mainfranken, Würzburg, Germany
| | - German Ott
- Department of Clinical Pathology, Robert-Bosch-Krankenhaus, Stuttgart, Germany.,Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
| | - Andreas Rosenwald
- Institute of Pathology, University of Würzburg and Comprehensive Cancer Center (CCC) Mainfranken, Würzburg, Germany
| |
Collapse
|
21
|
Puniya BL, Allen L, Hochfelder C, Majumder M, Helikar T. Systems Perturbation Analysis of a Large-Scale Signal Transduction Model Reveals Potentially Influential Candidates for Cancer Therapeutics. Front Bioeng Biotechnol 2016; 4:10. [PMID: 26904540 PMCID: PMC4750464 DOI: 10.3389/fbioe.2016.00010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 01/25/2016] [Indexed: 12/20/2022] Open
Abstract
Dysregulation in signal transduction pathways can lead to a variety of complex disorders, including cancer. Computational approaches such as network analysis are important tools to understand system dynamics as well as to identify critical components that could be further explored as therapeutic targets. Here, we performed perturbation analysis of a large-scale signal transduction model in extracellular environments that stimulate cell death, growth, motility, and quiescence. Each of the model’s components was perturbed under both loss-of-function and gain-of-function mutations. Using 1,300 simulations under both types of perturbations across various extracellular conditions, we identified the most and least influential components based on the magnitude of their influence on the rest of the system. Based on the premise that the most influential components might serve as better drug targets, we characterized them for biological functions, housekeeping genes, essential genes, and druggable proteins. The most influential components under all environmental conditions were enriched with several biological processes. The inositol pathway was found as most influential under inactivating perturbations, whereas the kinase and small lung cancer pathways were identified as the most influential under activating perturbations. The most influential components were enriched with essential genes and druggable proteins. Moreover, known cancer drug targets were also classified in influential components based on the affected components in the network. Additionally, the systemic perturbation analysis of the model revealed a network motif of most influential components which affect each other. Furthermore, our analysis predicted novel combinations of cancer drug targets with various effects on other most influential components. We found that the combinatorial perturbation consisting of PI3K inactivation and overactivation of IP3R1 can lead to increased activity levels of apoptosis-related components and tumor-suppressor genes, suggesting that this combinatorial perturbation may lead to a better target for decreasing cell proliferation and inducing apoptosis. Finally, our approach shows a potential to identify and prioritize therapeutic targets through systemic perturbation analysis of large-scale computational models of signal transduction. Although some components of the presented computational results have been validated against independent gene expression data sets, more laboratory experiments are warranted to more comprehensively validate the presented results.
Collapse
Affiliation(s)
- Bhanwar Lal Puniya
- Department of Biochemistry, University of Nebraska-Lincoln , Lincoln, NE , USA
| | - Laura Allen
- Department of Mathematics, University of Nebraska at Omaha , Omaha, NE , USA
| | | | - Mahbubul Majumder
- Department of Mathematics, University of Nebraska at Omaha , Omaha, NE , USA
| | - Tomáš Helikar
- Department of Biochemistry, University of Nebraska-Lincoln , Lincoln, NE , USA
| |
Collapse
|
22
|
Inhibition of DNA methyltransferase as a novel therapeutic strategy to overcome acquired resistance to dual PI3K/mTOR inhibitors. Oncotarget 2016; 6:5134-46. [PMID: 25762617 PMCID: PMC4467138 DOI: 10.18632/oncotarget.3016] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 12/31/2014] [Indexed: 12/24/2022] Open
Abstract
Dual PI3K/mTOR(phosphatidylinositol 3-kinase/mammalian target of rapamycin) inhibitors are being evaluated clinically for the treatment of tumors with a hyperactivated PI3K/mTOR pathway. However, unexpected outcomes were obtained in clinical studies of cancer patients with an aberrant PI3K pathway. In clinical trials, applicable combination regimens are not yet available. In this study, using an integrated analysis of acquired BEZ235-resistant nasopharyngeal carcinoma cells, we demonstrate that DNA methyltransferase is a key modulator and a common node upstream of the AKT/mTOR and PDK1/MYC pathways, which are activated in cancer cells with acquired BEZ235 resistance. DNA methyltransferases were upregulated and induced PTEN and PPP2R2B gene hypermethylation, which downregulated their expression in BEZ235-resistant cancer cells. Reduced PTEN and PPP2R2B expression correlated with activated AKT/mTOR and PDK1/MYC pathways and conferred considerable BEZ235 resistance in nasopharyngeal carcinoma. Targeting methyltransferases in combination with BEZ235 sensitized BEZ235-resistant cells to BEZ235 in vitro and in vivo, suggesting the potential clinical application of this strategy to overcome BEZ235 resistance.
Collapse
|
23
|
El Halabi L, Ghez D, Ribrag V. Novel targeted therapeutics for mantle cell lymphoma – What’s on the horizon? Expert Rev Hematol 2016; 9:271-81. [DOI: 10.1586/17474086.2016.1134309] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
24
|
Activity of BKM120 and BEZ235 against Lymphoma Cells. BIOMED RESEARCH INTERNATIONAL 2015; 2015:870918. [PMID: 26557706 PMCID: PMC4628710 DOI: 10.1155/2015/870918] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 07/27/2015] [Accepted: 08/19/2015] [Indexed: 12/23/2022]
Abstract
Non-Hodgkin lymphomas encompass a heterogeneous group of cancers, with 85–90% arising from B lymphocytes and the remainder deriving from T lymphocytes or NK lymphocytes. These tumors are molecularly and clinically heterogeneous, showing dramatically different responses and outcomes with standard therapies. Deregulated PI3K signaling is linked to oncogenesis and disease progression in hematologic malignancies and in a variety of solid tumors and apparently enhances resistance to antineoplastic therapy, resulting in a poor prognosis. Here, we have evaluated and compared the effects of the pan-PI3K inhibitor BKM120 and the dual PI3K/mTOR inhibitor BEZ235 on mantle, follicular, and T-cell lymphomas. Our results suggest that BKM120 and BEZ235 can effectively inhibit lymphoma cell proliferation by causing cell cycle arrest and can lead to cell death by inducing apoptosis and autophagy mediated by ROS accumulation. Despite great advances in lymphoma therapy after the introduction of monoclonal antibodies, many patients still die from disease progression. Therefore, novel treatment approaches are needed. BKM120 and BEZ235 alone and in combination are very effective against lymphoma cells in vitro. If further studies confirm their effectiveness in animal models, they may be promising candidates for development as new drugs.
Collapse
|
25
|
Lin Z, Mei H, Fan J, Yin Z, Wu G. Effect of the dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor NVP-BEZ235 against human Merkel cell carcinoma MKL-1 cells. Oncol Lett 2015; 10:3663-3667. [PMID: 26788188 DOI: 10.3892/ol.2015.3791] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Accepted: 05/22/2015] [Indexed: 12/29/2022] Open
Abstract
Merkel cell carcinoma (MCC) is an aggressive skin cancer with an increasing incidence. Aberrant activation of the phosphatidylinositol-3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway is common in human cancers and has been revealed to play an important function in cell proliferation, metabolism and tumorigenesis. In the present study, NVP-BEZ235, a dual PI3K/mTOR inhibitor, was revealed to be effective in inhibiting proliferation and inducing cell cycle arrest in MKL-1 cells. Additional investigations revealed that NVP-BEZ235 attenuated PI3K/Akt/mTOR signaling and upregulated the levels of the cell cycle inhibitors p21 and p27. Overall, the present results possess considerable implications for future development of dual PI3K/mTOR inhibitor as potential agents in the management of MCC.
Collapse
Affiliation(s)
- Zhenyu Lin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Hong Mei
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jiquan Fan
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Zhongyuan Yin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Gang Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
26
|
Zhi X, Chen W, Xue F, Liang C, Chen BW, Zhou Y, Wen L, Hu L, Shen J, Bai X, Liang T. OSI-027 inhibits pancreatic ductal adenocarcinoma cell proliferation and enhances the therapeutic effect of gemcitabine both in vitro and in vivo. Oncotarget 2015; 6:26230-41. [PMID: 26213847 PMCID: PMC4694897 DOI: 10.18632/oncotarget.4579] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 07/10/2015] [Indexed: 02/06/2023] Open
Abstract
Despite its relative rarity, pancreatic ductal adenocarcinoma (PDAC) accounts for a large percentage of cancer deaths. In this study, we investigated the in vitro efficacy of OSI-027, a selective inhibitor of mammalian target of rapamycin complex 1 (mTORC1) and mTORC2, to treat PDAC cell lines alone, and in combination with gemcitabine (GEM). Similarly, we tested the efficacy of these two compounds in a xenograft mouse model of PDAC. OSI-027 significantly arrested cell cycle in G0/G1 phase, inhibited the proliferation of Panc-1, BxPC-3, and CFPAC-1 cells, and downregulated mTORC1, mTORC2, phospho-Akt, phospho-p70S6K, phospho-4E-BP1, cyclin D1, and cyclin-dependent kinase 4 (CDK4) in these cells. Moreover, OSI-027 also downregulated multidrug resistance (MDR)-1, which has been implicated in chemotherapy resistance in PDAC cells and enhanced apoptosis induced by GEM in the three PDAC cell lines. When combined, OSI-027 with GEM showed synergistic cytotoxic effects both in vitro and in vivo. This is the first evidence of the efficacy of OSI-027 in PDAC and may provide the groundwork for a new clinical PDAC therapy.
Collapse
MESH Headings
- Animals
- Antimetabolites, Antineoplastic/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Apoptosis/drug effects
- Carcinoma, Pancreatic Ductal/drug therapy
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Cell Cycle Checkpoints
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cell Survival/drug effects
- Deoxycytidine/analogs & derivatives
- Deoxycytidine/pharmacology
- Dose-Response Relationship, Drug
- Drug Synergism
- Gene Expression Regulation, Neoplastic
- Humans
- Imidazoles/pharmacology
- Inhibitory Concentration 50
- Mechanistic Target of Rapamycin Complex 1
- Mechanistic Target of Rapamycin Complex 2
- Mice, Inbred BALB C
- Mice, Nude
- Multiprotein Complexes/antagonists & inhibitors
- Multiprotein Complexes/genetics
- Multiprotein Complexes/metabolism
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Protein Kinase Inhibitors/pharmacology
- RNA Interference
- Signal Transduction/drug effects
- TOR Serine-Threonine Kinases/antagonists & inhibitors
- TOR Serine-Threonine Kinases/genetics
- TOR Serine-Threonine Kinases/metabolism
- Time Factors
- Transfection
- Triazines/pharmacology
- Tumor Burden/drug effects
- Xenograft Model Antitumor Assays
- Gemcitabine
Collapse
Affiliation(s)
- Xiao Zhi
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P.R.China
| | - Wei Chen
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P.R.China
| | - Fei Xue
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P.R.China
| | - Chao Liang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P.R.China
| | - Bryan Wei Chen
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P.R.China
| | - Yue Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P.R.China
| | - Liang Wen
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P.R.China
| | - Liqiang Hu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P.R.China
| | - Jian Shen
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P.R.China
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P.R.China
- Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P.R.China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P.R.China
- Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P.R.China
- Collaborative Innovation Center for Cancer Medicine, Zhejiang University, Hangzhou, P.R.China
| |
Collapse
|
27
|
Wang H, Huang F, Wang J, Wang P, Lv W, Hong L, Li S, Zhou J. The synergistic inhibition of breast cancer proliferation by combined treatment with 4EGI-1 and MK2206. Cell Cycle 2015; 14:232-42. [PMID: 25607647 DOI: 10.4161/15384101.2014.977096] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cap-dependent translation is a potential cancer-related target (oncotarget) due to its critical role in cancer initiation and progression. 4EGI-1, an inhibitor of eIF4E/eIF4G interaction, was discovered by screening chemical libraries of small molecules. 4EGI-1 inhibits cap-dependent translation initiation by impairing the assembly of the eIF4E/eIF4G complex, and therefore is a potential anti-cancer agent. Here, we report that 4EGI-1 also inhibits mTORC1 signaling independent of its inhibitory role on cap-dependent translation initiation. The inhibition of mTORC1 signaling by 4EGI-1 activates Akt due to both abrogation of the negative feedback loops from mTORC1 to PI3K and activation of mTORC2. We further validated that mTORC2 activity is required for 4EGI-1-mediated Akt activation. The activated Akt counteracted the anticancer effects of 4EGI-1. In support of this model, inhibition of Akt potentiates the antitumor activity of 4EGI-1 both in vitro and in a xenograft mouse model in vivo. Our results suggest that a combination of 4EGI-1and Akt inhibitor is a rational approach for the treatment of cancer.
Collapse
Affiliation(s)
- Hongtao Wang
- a Laboratory of Medical Molecular Biology; Beijing Institute of Biotechnology ; Beijing , P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Sisakhtnezhad S, Khosravi L. Emerging physiological and pathological implications of tunneling nanotubes formation between cells. Eur J Cell Biol 2015; 94:429-43. [PMID: 26164368 DOI: 10.1016/j.ejcb.2015.06.010] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 06/20/2015] [Accepted: 06/23/2015] [Indexed: 12/21/2022] Open
Abstract
Cell-to-cell communication is a critical requirement to coordinate behaviors of the cells in a community and thereby achieve tissue homeostasis and conservation of the multicellular organisms. Tunneling nanotubes (TNTs), as a cell-to-cell communication over long distance, allow for bi- or uni-directional transfer of cellular components between cells. Identification of inducing agents and the cell and molecular mechanism underling the formation of TNTs and their structural and functional features may lead to finding new important roles for these intercellular bridges in vivo and in vitro. During the last decade, research has shown TNTs have different structural and functional properties, varying between and within cell systems. In this review, we will focus on TNTs and their cell and molecular mechanism of formation. Moreover, the latest findings into their functional roles in physiological and pathological processes, such as signal transduction, micro and nano-particles delivery, immune responses, embryogenesis, cellular reprogramming, apoptosis, cancer, and neurodegenerative diseases initiation and progression and pathogens transfer, will be discussed.
Collapse
Affiliation(s)
| | - Leila Khosravi
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| |
Collapse
|
29
|
Yang H, Chen H, Guo H, Li W, Tang J, Xu B, Sun M, Ding G, Jiang L, Cui D, Zheng X, Duan Y. Molecular mechanisms of 2, 3', 4, 4', 5-pentachlorobiphenyl-induced thyroid dysfunction in FRTL-5 cells. PLoS One 2015; 10:e0120133. [PMID: 25789747 PMCID: PMC4366388 DOI: 10.1371/journal.pone.0120133] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 01/19/2015] [Indexed: 12/02/2022] Open
Abstract
Polychlorinated biphenyls (PCBs) can severely interfere with multiple animals and human systems. To explore the molecular mechanisms underlying 2, 3′, 4, 4′, 5- pentachlorobiphenyl (PCB118)-induced thyroid dysfunction, Fischer rat thyroid cell line-5(FRTL-5) cells were treated with either different concentrations of PCB118 or dimethyl sulfoxide (DMSO). The effects of PCB118 on FRTL-5 cells viability and apoptosis were assessed by using a Cell Counting Kit-8 assay and apoptosis assays, respectively. Quantitative real-time polymerase chain reaction was used to quantify protein kinase B (Akt), Forkhead box protein O3a (FoxO3a), and sodium/iodide symporter (NIS) mRNA expression levels. Western blotting was used to detect Akt, phospho-Akt (p-Akt), FoxO3a, phospho-FoxO3a (p-FoxO3a), and NIS protein levels. Luciferase reporter gene technology was used to detect the transcriptional activities of FoxO3a and NIS promoters. The effects of the constitutively active Akt (CA-Akt) and dominant-negative Akt (DN-Akt) plasmids on p-Akt, p-FoxO3a, and NIS levels were examined in PCB118-treated FRTL-5 cells. The effects of FoxO3a siRNA on FoxO3a, p-FoxO3a, and NIS protein levels were examined in the PCB118-treated FRTL-5 cells. The effects of pcDNA3 (plsmid vectors designed for high-level stable and transient expression in mammalian host)-FoxO3a on NIS promoter activity were examined in the PCB118-treated FRTL-5 cells. Our results indicated that relatively higher PCB118 concentrations can inhibit cell viability in a concentration- and time-dependent manner. Akt, p-Akt, and p-FoxO3a protein or mRNA levels increased significantly in PCB118-treated groups and NIS protein and mRNA levels decreased considerably compared with the control groups. FoxO3a promoter activity increased significantly, whereas NIS promoter activity decreased. These effects on p-FoxO3a and NIS could be decreased by the DN-Akt plasmid, enhanced by the CA-Akt plasmid, and blocked by FoxO3a siRNA. The overexpressed FoxO3a could reduce NIS promoter activity. Our results suggested that PCB118 induces thyroid cell dysfunction through the Akt/FoxO3a/NIS signaling pathway.
Collapse
Affiliation(s)
- Hui Yang
- Department of Endocrinology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Huanhuan Chen
- Department of Endocrinology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hongwei Guo
- Department of Endocrinology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wen Li
- Department of Endocrinology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jinmei Tang
- Department of Endocrinology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Bojin Xu
- Department of Endocrinology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Minne Sun
- Department of Endocrinology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Guoxian Ding
- Department of Gerontology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lin Jiang
- Department of Endocrinology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Dai Cui
- Department of Endocrinology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xuqin Zheng
- Department of Endocrinology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yu Duan
- Department of Endocrinology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- * E-mail:
| |
Collapse
|
30
|
Xiao Z, Ni Y, Yin G, Wu H, Li J, Miao K. Mantle cell lymphoma concurrent with T-large granular lymphocytic leukemia: report of a case and review of literature. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:3365-3369. [PMID: 26045870 PMCID: PMC4440179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Accepted: 02/23/2015] [Indexed: 06/04/2023]
Abstract
Mantle cell lymphoma is one of the B-cell lymphomas. The concurrent presentation of mantle cell lymphoma with large granular lymphocytic leukemia simultaneously has never been reported. In this case we present an old man with concomitant mantle cell lymphoma and large granular lymphocytic leukemia diagnosed by the morphology of the bone marrow aspiration, immunophenotyping of the peripheral blood by flow cytometry detecting the increased CD3+CD4-CD8+ cells, immunohistochemical studies of lymph node showed cyclinD1+, chromosome analysis by fluorescence in situ hybridization (FISH) showed t(11,14), positive results of IGH and TCR rearrangement studies. The patient discharged from the hospital voluntarily and lost the follow-up. A brief discussion is also presented.
Collapse
MESH Headings
- Aged
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/genetics
- Bone Marrow Examination
- Chromosomes, Human, Pair 11
- Chromosomes, Human, Pair 14
- Flow Cytometry
- Gene Rearrangement
- Gene Rearrangement, T-Lymphocyte
- Genes, Immunoglobulin Heavy Chain
- Genetic Predisposition to Disease
- Humans
- Immunohistochemistry
- Immunophenotyping
- In Situ Hybridization, Fluorescence
- Leukemia, Large Granular Lymphocytic/genetics
- Leukemia, Large Granular Lymphocytic/immunology
- Leukemia, Large Granular Lymphocytic/pathology
- Lymphoma, Mantle-Cell/genetics
- Lymphoma, Mantle-Cell/immunology
- Lymphoma, Mantle-Cell/pathology
- Male
- Neoplasms, Multiple Primary/genetics
- Neoplasms, Multiple Primary/immunology
- Neoplasms, Multiple Primary/pathology
- Phenotype
- Translocation, Genetic
Collapse
Affiliation(s)
- Zhengrui Xiao
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital Nanjing 210029, China
| | - Ying Ni
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital Nanjing 210029, China
| | - Guangli Yin
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital Nanjing 210029, China
| | - Hanxin Wu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital Nanjing 210029, China
| | - Jianyong Li
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital Nanjing 210029, China
| | - Kourong Miao
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital Nanjing 210029, China
| |
Collapse
|
31
|
van Krieken JH. New developments in the pathology of malignant lymphoma. A review of the literature published from August 2014 to October 2014. J Hematop 2014. [DOI: 10.1007/s12308-014-0224-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|