201
|
Xu ZZ, Shen JK, Zhao SQ, Li JM. Clinical significance of chemokine receptor CXCR4 and mammalian target of rapamycin (mTOR) expression in patients with diffuse large B-cell lymphoma. Leuk Lymphoma 2017; 59:1451-1460. [PMID: 28952842 DOI: 10.1080/10428194.2017.1379077] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
To assess the relevance of C-X-C chemokine receptor type 4 (CXCR4) and mammalian target of rapamycin (mTOR) to large-B-cell lymphoma (DLBCL), levels of protein expression were measured in 56 DLBCL patients who had received rituximab-based therapy. Of these, 34 were positive for CXCR4 expression (60.7%) and 31 for mTOR (55.4%). CXCR4 expression was positively correlated with mTOR expression (r = 0.602; p = .000). CXCR4 expression was significantly associated with high lactate dehydrogenase (LDH) level (p = .009), high IPI score (p = .030) and non-GCB subtype (p = .006). Furthermore, the expression levels of CXCR4 and mTOR were negatively correlated with the chance of remission (p < .05). Kaplan-Meier analysis indicated significantly shorter progression-free survival (PFS) and overall survival (OS) in patients positive for CXCR4 and mTOR expression. The combination therapy with CXCR4 inhibitor WZ811 and mTOR inhibitor everolimus showed syncergistic effect in DLBCL cell lines. These results suggest that the expression of CXCR4 and mTOR may be suitable as biomarkers of the prognosis of DLBCL and for development of new therapeutic strategies.
Collapse
Affiliation(s)
- Zi-Zhen Xu
- a Department of Laboratory Medicine , Ruijin Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Jian-Kang Shen
- b Department of Surgery , Luwan Branch of Ruijin Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Shu-Qing Zhao
- c Department of Hematology , Luwan Branch of Ruijin Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Jun-Min Li
- d Department of Hematology , Ruijin Hospital, Shanghai Jiao-Tong University School of Medicine , Shanghai , China
| |
Collapse
|
202
|
Pongas GN, Annunziata CM, Staudt LM. PI3Kδ inhibition causes feedback activation of PI3Kα in the ABC subtype of diffuse large B-cell lymphoma. Oncotarget 2017; 8:81794-81802. [PMID: 29137222 PMCID: PMC5669848 DOI: 10.18632/oncotarget.20864] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 08/15/2017] [Indexed: 11/25/2022] Open
Abstract
Cell line models of the activated B cell-like (ABC) subtype of diffuse large B cell (DLBCL) depend on both NF-κB and phosphatidylinositol 3-kinase (PI3K) signaling pathways for survival, especially those with gain-of-function B cell receptor (BCR) mutations. Here we show that these cells depend specifically on the PI3Kδ isoform, but that PI3K pathway interruption by PI3Kδ inhibitors is short-lived due to feedback activation of the PI3Kα isoform. PI3Kδ and PI3Kα inhibition cooperated in killing ABC DLBCL lines, and genetic knockdown of PI3Kα sensitized cells to PI3Kδ inhibition and prolonged the interruption of PI3K signaling. PI3Kδ inhibition evoked feedback activation of proximal BCR signaling, which increased the association of PI3Kα with BCAP and CD19 and increased overall PI3K activity. These results support the clinical evaluation of dual PI3Kδ and PI3Kα inhibition in patients with ABC DLBCL.
Collapse
Affiliation(s)
- Georgios N Pongas
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.,Medical Oncology Service, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.,Women's Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Christina M Annunziata
- Medical Oncology Service, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.,Women's Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Louis M Staudt
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
203
|
Heske CM, Davis MI, Baumgart JT, Wilson K, Gormally MV, Chen L, Zhang X, Ceribelli M, Duveau DY, Guha R, Ferrer M, Arnaldez FI, Ji J, Tran HL, Zhang Y, Mendoza A, Helman LJ, Thomas CJ. Matrix Screen Identifies Synergistic Combination of PARP Inhibitors and Nicotinamide Phosphoribosyltransferase (NAMPT) Inhibitors in Ewing Sarcoma. Clin Cancer Res 2017; 23:7301-7311. [PMID: 28899971 DOI: 10.1158/1078-0432.ccr-17-1121] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 08/04/2017] [Accepted: 09/07/2017] [Indexed: 12/20/2022]
Abstract
Purpose: Although many cancers are showing remarkable responses to targeted therapies, pediatric sarcomas, including Ewing sarcoma, remain recalcitrant. To broaden the therapeutic landscape, we explored the in vitro response of Ewing sarcoma cell lines against a large collection of investigational and approved drugs to identify candidate combinations.Experimental Design: Drugs displaying activity as single agents were evaluated in combinatorial (matrix) format to identify highly active, synergistic drug combinations, and combinations were subsequently validated in multiple cell lines using various agents from each class. Comprehensive metabolomic and proteomic profiling was performed to better understand the mechanism underlying the synergy. Xenograft experiments were performed to determine efficacy and in vivo mechanism.Results: Several promising candidates emerged, including the combination of small-molecule PARP and nicotinamide phosphoribosyltransferase (NAMPT) inhibitors, a rational combination as NAMPTis block the rate-limiting enzyme in the production of nicotinamide adenine dinucleotide (NAD+), a necessary substrate of PARP. Mechanistic drivers of the synergistic cell killing phenotype of these combined drugs included depletion of NMN and NAD+, diminished PAR activity, increased DNA damage, and apoptosis. Combination PARPis and NAMPTis in vivo resulted in tumor regression, delayed disease progression, and increased survival.Conclusions: These studies highlight the potential of these drugs as a possible therapeutic option in treating patients with Ewing sarcoma. Clin Cancer Res; 23(23); 7301-11. ©2017 AACR.
Collapse
Affiliation(s)
- Christine M Heske
- Molecular Oncology Section, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| | - Mindy I Davis
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Joshua T Baumgart
- Molecular Oncology Section, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Kelli Wilson
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Michael V Gormally
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Lu Chen
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Xiaohu Zhang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Michele Ceribelli
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Damien Y Duveau
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Rajarshi Guha
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Marc Ferrer
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Fernanda I Arnaldez
- Molecular Oncology Section, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jiuping Ji
- National Clinical Target Validation Laboratory, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Huong-Lan Tran
- National Clinical Target Validation Laboratory, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Yiping Zhang
- National Clinical Target Validation Laboratory, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Arnulfo Mendoza
- Molecular Oncology Section, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Lee J Helman
- Molecular Oncology Section, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Craig J Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland.
| |
Collapse
|
204
|
Havranek O, Xu J, Köhrer S, Wang Z, Becker L, Comer JM, Henderson J, Ma W, Man Chun Ma J, Westin JR, Ghosh D, Shinners N, Sun L, Yi AF, Karri AR, Burger JA, Zal T, Davis RE. Tonic B-cell receptor signaling in diffuse large B-cell lymphoma. Blood 2017; 130:995-1006. [PMID: 28646116 PMCID: PMC5813722 DOI: 10.1182/blood-2016-10-747303] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 06/16/2017] [Indexed: 12/14/2022] Open
Abstract
We used clustered regularly interspaced short palindromic repeats/Cas9-mediated genomic modification to investigate B-cell receptor (BCR) signaling in cell lines of diffuse large B-cell lymphoma (DLBCL). Three manipulations that altered BCR genes without affecting surface BCR levels showed that BCR signaling differs between the germinal center B-cell (GCB) subtype, which is insensitive to Bruton tyrosine kinase inhibition by ibrutinib, and the activated B-cell (ABC) subtype. Replacing antigen-binding BCR regions had no effect on BCR signaling in GCB-DLBCL lines, reflecting this subtype's exclusive use of tonic BCR signaling. Conversely, Y188F mutation in the immunoreceptor tyrosine-based activation motif of CD79A inhibited tonic BCR signaling in GCB-DLBCL lines but did not affect their calcium flux after BCR cross-linking or the proliferation of otherwise-unmodified ABC-DLBCL lines. CD79A-GFP fusion showed BCR clustering or diffuse distribution, respectively, in lines of ABC and GCB subtypes. Tonic BCR signaling acts principally to activate AKT, and forced activation of AKT rescued GCB-DLBCL lines from knockout (KO) of the BCR or 2 mediators of tonic BCR signaling, SYK and CD19. The magnitude and importance of tonic BCR signaling to proliferation and size of GCB-DLBCL lines, shown by the effect of BCR KO, was highly variable; in contrast, pan-AKT KO was uniformly toxic. This discrepancy was explained by finding that BCR KO-induced changes in AKT activity (measured by gene expression, CXCR4 level, and a fluorescent reporter) correlated with changes in proliferation and with baseline BCR surface density. PTEN protein expression and BCR surface density may influence clinical response to therapeutic inhibition of tonic BCR signaling in DLBCL.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - R Eric Davis
- Department of Lymphoma and Myeloma
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
205
|
Alileche A, Hampikian G. The effect of Nullomer-derived peptides 9R, 9S1R and 124R on the NCI-60 panel and normal cell lines. BMC Cancer 2017; 17:533. [PMID: 28793867 PMCID: PMC5551024 DOI: 10.1186/s12885-017-3514-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 07/28/2017] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Nullomer peptides are the smallest sequences absent from databases of natural proteins. We first began compiling a list of absent 5-amino acid strings in 2006 (1). We report here the effects of Nullomer-derived peptides 9R, 9S1R and 124R on the NCI-60 panel, derived from human cancers of 9 organs (kidney, ovary, skin melanoma, lung, brain, lung, colon, prostate and the hematopoietic system), and four normal cell lines (endothelial HUVEC, skin fibroblasts BJ, colon epithelial FHC and normal prostate RWPE-1). METHODS NCI-60 cancer cell panel and four normal cell lines were cultured in vitro in RPMI1640 supplemented with 10% Hyclone fetal bovine serum and exposed for 48 h to 5 μM, 25 μM and 50 μM of peptides 9R, 9S1R and 124R. Viability was assessed by CCK-8 assay. For peptide ATP depletion effects, one cell line representing each organ in the NCI-60 panel, and four normal cell lines were exposed to 50 μM of peptides 9R, 9S1R and 124R for 3 h. The ATP content was assessed in whole cells, and their supernatants. RESULTS Peptides 9S1R and 9R are respectively lethal to 95 and 81.6% of the 60 cancer cell lines tested. Control peptide 124R has no effect on the growth of these cells. Especially interesting the fact that peptides 9R and 9S1R are capable of killing drug-resistant and hormone-resistant cell lines, and even cancer stem cells. Peptides 9R and 9S1R have a broader activity spectrum than many cancer drugs in current use, can completely deplete cellular ATP within 3 h, and are less toxic to 3 of the 4 normal cell lines tested than they are to several cancers. CONCLUSIONS Nullomer peptides 9R and 9S1R have a large broad lethal effect on cancer cell lines derived from nine organs represented in the NCI-60 panel. This broad activity crosses many of the categorical divisions used in the general classification of cancers: solid vs liquid cancers, drug sensitive vs drug resistant, hormone sensitive vs hormone resistant, cytokine sensitive vs cytokine non sensitive, slow growing vs rapid growing, differentiated vs dedifferentiated cancers. Furthermore peptides 9R and 9S1R are lethal to cancer stem cells and breast canrcinosarcoma.
Collapse
Affiliation(s)
- Abdelkrim Alileche
- Biology Department Room SN-215, Boise State University, 1910 University Drive, Boise, ID 83725 USA
| | - Greg Hampikian
- Biology Department Room SN-215, Boise State University, 1910 University Drive, Boise, ID 83725 USA
| |
Collapse
|
206
|
Beretta GL, Cassinelli G, Pennati M, Zuco V, Gatti L. Overcoming ABC transporter-mediated multidrug resistance: The dual role of tyrosine kinase inhibitors as multitargeting agents. Eur J Med Chem 2017; 142:271-289. [PMID: 28851502 DOI: 10.1016/j.ejmech.2017.07.062] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 07/21/2017] [Accepted: 07/25/2017] [Indexed: 12/14/2022]
Abstract
Resistance to conventional and target specific antitumor drugs still remains one of the major cause of treatment failure and patience death. This condition often involves ATP-binding cassette (ABC) transporters that, by pumping the drugs outside from cancer cells, attenuate the potency of chemotherapeutics and negatively impact on the fate of anticancer therapy. In recent years, several tyrosine kinase inhibitors (TKIs) (e.g., imatinib, nilotinib, dasatinib, ponatinib, gefitinib, erlotinib, lapatinib, vandetanib, sunitinib, sorafenib) have been reported to interact with ABC transporters (e.g., ABCB1, ABCC1, ABCG2, ABCC10). This finding disclosed a very complex scenario in which TKIs may behave as substrates or inhibitors depending on the expression of specific pumps, drug concentration, affinity for transporters and types of co-administered agents. In this context, in-depth investigation on TKI chemosensitizing functions might provide a strong rationale for combining TKIs and conventional therapeutics in specific malignancies. The reposition of TKIs as antagonists of ABC transporters opens a new way towards anticancer therapy and clinical strategies aimed at counteracting drug resistance. This review will focus on some paradigmatic examples of the complex and not yet fully elucidated interaction between clinical available TKIs (e.g. BCR-ABL, EGFR, VEGFR inhibitors) with the main ABC transporters implicated in multidrug resistance.
Collapse
Affiliation(s)
- Giovanni Luca Beretta
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milano, Italy.
| | - Giuliana Cassinelli
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milano, Italy.
| | - Marzia Pennati
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milano, Italy.
| | - Valentina Zuco
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milano, Italy.
| | - Laura Gatti
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milano, Italy.
| |
Collapse
|
207
|
Schaffer M, Chaturvedi S, Davis C, Aquino R, Stepanchick E, Versele M, Liu Y, Yang J, Lu R, Balasubramanian S. Identification of potential ibrutinib combinations in hematological malignancies using a combination high-throughput screen. Leuk Lymphoma 2017; 59:931-940. [DOI: 10.1080/10428194.2017.1349899] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Michael Schaffer
- Oncology, Translational Research, Janssen Pharmaceutical Companies of Johnson & Johnson, Spring House, PA, USA
| | - Shalini Chaturvedi
- Oncology, Translational Research, Janssen Pharmaceutical Companies of Johnson & Johnson, Spring House, PA, USA
| | - Cuc Davis
- Oncology, Translational Research, Janssen Pharmaceutical Companies of Johnson & Johnson, Spring House, PA, USA
| | - Regina Aquino
- Oncology, Translational Research, Janssen Pharmaceutical Companies of Johnson & Johnson, Spring House, PA, USA
| | - Emily Stepanchick
- Oncology, Translational Research, Janssen Pharmaceutical Companies of Johnson & Johnson, Spring House, PA, USA
| | | | - Yang Liu
- Janssen China Research & Development, Shanghai, China
| | - Jennifer Yang
- Janssen China Research & Development, Shanghai, China
| | - Rongzhen Lu
- Janssen China Research & Development, Shanghai, China
| | - Sriram Balasubramanian
- Oncology, Translational Research, Janssen Pharmaceutical Companies of Johnson & Johnson, Spring House, PA, USA
| |
Collapse
|
208
|
Dermit M, Dokal A, Cutillas PR. Approaches to identify kinase dependencies in cancer signalling networks. FEBS Lett 2017; 591:2577-2592. [DOI: 10.1002/1873-3468.12748] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 06/27/2017] [Accepted: 07/03/2017] [Indexed: 12/18/2022]
Affiliation(s)
- Maria Dermit
- Cell Signalling & Proteomics Group; Barts Cancer Institute (CRUK Centre); Queen Mary University of London; UK
| | - Arran Dokal
- Cell Signalling & Proteomics Group; Barts Cancer Institute (CRUK Centre); Queen Mary University of London; UK
| | - Pedro R. Cutillas
- Cell Signalling & Proteomics Group; Barts Cancer Institute (CRUK Centre); Queen Mary University of London; UK
| |
Collapse
|
209
|
Sensitivity to PI3K and AKT inhibitors is mediated by divergent molecular mechanisms in subtypes of DLBCL. Blood 2017; 130:310-322. [DOI: 10.1182/blood-2016-12-758599] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 02/10/2017] [Indexed: 01/14/2023] Open
Abstract
Key Points
PI3Kα/δ inhibition induces cytotoxicity in ABC DLBCLs through downregulation of NF-κB signaling. Inhibition of AKT induces cytotoxicity by downregulation of MYC in PTEN-deficient DLBCL models in vivo and in vitro.
Collapse
|
210
|
Ono A, Sano O, Kazetani KI, Muraki T, Imamura K, Sumi H, Matsui J, Iwata H. Feedback activation of AMPK-mediated autophagy acceleration is a key resistance mechanism against SCD1 inhibitor-induced cell growth inhibition. PLoS One 2017; 12:e0181243. [PMID: 28704514 PMCID: PMC5509324 DOI: 10.1371/journal.pone.0181243] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 06/28/2017] [Indexed: 01/08/2023] Open
Abstract
Elucidating the bioactive compound modes of action is crucial for increasing success rates in drug development. For anticancer drugs, defining effective drug combinations that overcome resistance improves therapeutic efficacy. Herein, by using a biologically annotated compound library, we performed a large-scale combination screening with Stearoyl-CoA desaturase-1 (SCD1) inhibitor, T-3764518, which partially inhibits colorectal cancer cell proliferation. T-3764518 induced phosphorylation and activation of AMPK in HCT-116 cells, which led to blockade of downstream fatty acid synthesis and acceleration of autophagy. Attenuation of fatty acid synthesis by small molecules suppressed the growth inhibitory effect of T-3764518. In contrast, combination of T-3764518 with autophagy flux inhibitors synergistically inhibited cellular proliferation. Experiments using SCD1 knock-out cells validated the results obtained with T-3764518. The results of our study indicated that activation of autophagy serves as a survival signal when SCD1 is inhibited in HCT-116 cells. Furthermore, these findings suggest that combining SCD1 inhibitor with autophagy inhibitors is a promising anticancer therapy.
Collapse
Affiliation(s)
- Akito Ono
- Biomolecular Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Ltd., Fujisawa, Kanagawa, Japan
| | - Osamu Sano
- Biomolecular Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Ltd., Fujisawa, Kanagawa, Japan
| | - Ken-ichi Kazetani
- Biomolecular Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Ltd., Fujisawa, Kanagawa, Japan
| | - Takamichi Muraki
- Biomolecular Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Ltd., Fujisawa, Kanagawa, Japan
| | - Keisuke Imamura
- Oncology Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Ltd., Fujisawa, Kanagawa, Japan
| | - Hiroyuki Sumi
- Oncology Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Ltd., Fujisawa, Kanagawa, Japan
| | - Junji Matsui
- Biomolecular Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Ltd., Fujisawa, Kanagawa, Japan
| | - Hidehisa Iwata
- Biomolecular Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Ltd., Fujisawa, Kanagawa, Japan
- * E-mail:
| |
Collapse
|
211
|
Abstract
In this review, we summarize recent work exploring a novel conceptual approach termed "synthetic essentiality" as a means for targeting specific tumor suppressor gene deficiencies in cancer. With the aid of extensive publically available cancer genome and clinical databases, "synthetic essentiality" could be utilized to identify synthetic essential genes, which might be occasionally deleted in some cancers but almost always retained in the context of a specific tumor suppressor deficiency. Synthetic essentiality expands the existing concepts for therapeutic strategies, including oncogene addiction, tumor maintenance, synthetic, and collateral lethality, to provide a framework for the discovery of cancer-specific vulnerabilities. Enabled by ever-expanding large-scale genome datasets and genome-scale functional screens, the "synthetic essentiality" framework provides an avenue for the identification of context-specific therapeutic targets and development of patient responder hypotheses for novel and existing therapies.
Collapse
Affiliation(s)
- Di Zhao
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ronald A DePinho
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
212
|
Holbeck SL, Camalier R, Crowell JA, Govindharajulu JP, Hollingshead M, Anderson LW, Polley E, Rubinstein L, Srivastava A, Wilsker D, Collins JM, Doroshow JH. The National Cancer Institute ALMANAC: A Comprehensive Screening Resource for the Detection of Anticancer Drug Pairs with Enhanced Therapeutic Activity. Cancer Res 2017; 77:3564-3576. [PMID: 28446463 PMCID: PMC5499996 DOI: 10.1158/0008-5472.can-17-0489] [Citation(s) in RCA: 193] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/13/2017] [Accepted: 04/24/2017] [Indexed: 12/22/2022]
Abstract
To date, over 100 small-molecule oncology drugs have been approved by the FDA. Because of the inherent heterogeneity of tumors, these small molecules are often administered in combination to prevent emergence of resistant cell subpopulations. Therefore, new combination strategies to overcome drug resistance in patients with advanced cancer are needed. In this study, we performed a systematic evaluation of the therapeutic activity of over 5,000 pairs of FDA-approved cancer drugs against a panel of 60 well-characterized human tumor cell lines (NCI-60) to uncover combinations with greater than additive growth-inhibitory activity. Screening results were compiled into a database, termed the NCI-ALMANAC (A Large Matrix of Anti-Neoplastic Agent Combinations), publicly available at https://dtp.cancer.gov/ncialmanac Subsequent in vivo experiments in mouse xenograft models of human cancer confirmed combinations with greater than single-agent efficacy. Concomitant detection of mechanistic biomarkers for these combinations in vivo supported the initiation of two phase I clinical trials at the NCI to evaluate clofarabine with bortezomib and nilotinib with paclitaxel in patients with advanced cancer. Consequently, the hypothesis-generating NCI-ALMANAC web-based resource has demonstrated value in identifying promising combinations of approved drugs with potent anticancer activity for further mechanistic study and translation to clinical trials. Cancer Res; 77(13); 3564-76. ©2017 AACR.
Collapse
Affiliation(s)
- Susan L Holbeck
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, Maryland
| | - Richard Camalier
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, Maryland
| | - James A Crowell
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, Maryland
| | - Jeevan Prasaad Govindharajulu
- Clinical Pharmacodynamics Program, Applied/Developmental Research Directorate, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Melinda Hollingshead
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, Maryland
| | - Lawrence W Anderson
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, Maryland
| | - Eric Polley
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, Maryland
| | - Larry Rubinstein
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, Maryland
| | - Apurva Srivastava
- Clinical Pharmacodynamics Program, Applied/Developmental Research Directorate, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Deborah Wilsker
- Clinical Pharmacodynamics Program, Applied/Developmental Research Directorate, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Jerry M Collins
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, Maryland
| | - James H Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, Maryland.
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| |
Collapse
|
213
|
Niemann CU, Mora-Jensen HI, Dadashian EL, Krantz F, Covey T, Chen SS, Chiorazzi N, Izumi R, Ulrich R, Lannutti BJ, Wiestner A, Herman SEM. Combined BTK and PI3Kδ Inhibition with Acalabrutinib and ACP-319 Improves Survival and Tumor Control in CLL Mouse Model. Clin Cancer Res 2017. [PMID: 28645939 DOI: 10.1158/1078-0432.ccr-17-0650] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Purpose: Targeting the B-cell receptor (BCR) pathway with inhibitors of Bruton tyrosine kinase (BTK) and PI3Kδ is highly effective for the treatment of chronic lymphocytic leukemia (CLL). However, deep remissions are uncommon, and drug resistance with single-agent therapy can occur. In vitro studies support the effectiveness of combing PI3Kδ and BTK inhibitors.Experimental Design: As CLL proliferation and survival depends on the microenvironment, we used murine models to assess the efficacy of the BTK inhibitor acalabrutinib combined with the PI3Kδ inhibitor ACP-319 in vivo We compared single-agent with combination therapy in TCL1-192 cell-injected mice, a model of aggressive CLL.Results: We found significantly larger reductions in tumor burden in the peripheral blood and spleen of combination-treated mice. Although single-agent therapy improved survival compared with control mice by a few days, combination therapy extended survival by over 2 weeks compared with either single agent. The combination reduced tumor proliferation, NF-κB signaling, and expression of BCL-xL and MCL-1 more potently than single-agent therapy.Conclusions: The combination of acalabrutinib and ACP-319 was superior to single-agent treatment in a murine CLL model, warranting further investigation of this combination in clinical studies. Clin Cancer Res; 23(19); 5814-23. ©2017 AACR.
Collapse
Affiliation(s)
- Carsten U Niemann
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland.,Department of Hematology, Rigshospitalet, Copenhagen, Denmark
| | - Helena I Mora-Jensen
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Eman L Dadashian
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | | | | | - Shih-Shih Chen
- The Feinstein Institute for Medical Research, Northwell Health, Manhasset, New York
| | - Nicholas Chiorazzi
- The Feinstein Institute for Medical Research, Northwell Health, Manhasset, New York.,Department of Medicine, Hofstra Northwell School of Medicine, Hempstead, New York.,Department of Molecular Medicine, Hofstra Northwell School of Medicine, Hempstead, New York
| | | | | | | | - Adrian Wiestner
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland.
| | - Sarah E M Herman
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland.
| |
Collapse
|
214
|
Lionakis MS, Dunleavy K, Roschewski M, Widemann BC, Butman JA, Schmitz R, Yang Y, Cole DE, Melani C, Higham CS, Desai JV, Ceribelli M, Chen L, Thomas CJ, Little RF, Gea-Banacloche J, Bhaumik S, Stetler-Stevenson M, Pittaluga S, Jaffe ES, Heiss J, Lucas N, Steinberg SM, Staudt LM, Wilson WH. Inhibition of B Cell Receptor Signaling by Ibrutinib in Primary CNS Lymphoma. Cancer Cell 2017; 31:833-843.e5. [PMID: 28552327 PMCID: PMC5571650 DOI: 10.1016/j.ccell.2017.04.012] [Citation(s) in RCA: 384] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Revised: 03/09/2017] [Accepted: 04/18/2017] [Indexed: 01/11/2023]
Abstract
Primary CNS lymphoma (PCNSL) harbors mutations that reinforce B cell receptor (BCR) signaling. Ibrutinib, a Bruton's tyrosine kinase (BTK) inhibitor, targets BCR signaling and is particularly active in lymphomas with mutations altering the BCR subunit CD79B and MYD88. We performed a proof-of-concept phase Ib study of ibrutinib monotherapy followed by ibrutinib plus chemotherapy (DA-TEDDi-R). In 18 PCNSL patients, 94% showed tumor reductions with ibrutinib alone, including patients having PCNSL with CD79B and/or MYD88 mutations, and 86% of evaluable patients achieved complete remission with DA-TEDDi-R. Increased aspergillosis was observed with ibrutinib monotherapy and DA-TEDDi-R. Aspergillosis was linked to BTK-dependent fungal immunity in a murine model. PCNSL is highly dependent on BCR signaling, and ibrutinib appears to enhance the efficacy of chemotherapy.
Collapse
Affiliation(s)
- Michail S Lionakis
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kieron Dunleavy
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mark Roschewski
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Brigitte C Widemann
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - John A Butman
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Roland Schmitz
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yandan Yang
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Diane E Cole
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christopher Melani
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christine S Higham
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jigar V Desai
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michele Ceribelli
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Gaithersburg, MD 20850, USA
| | - Lu Chen
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Gaithersburg, MD 20850, USA
| | - Craig J Thomas
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Gaithersburg, MD 20850, USA
| | - Richard F Little
- Cancer Therapy Evaluation Program, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Juan Gea-Banacloche
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sucharita Bhaumik
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Maryalice Stetler-Stevenson
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Stefania Pittaluga
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Elaine S Jaffe
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - John Heiss
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicole Lucas
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Seth M Steinberg
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Louis M Staudt
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Wyndham H Wilson
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
215
|
Control of B lymphocyte development and functions by the mTOR signaling pathways. Cytokine Growth Factor Rev 2017; 35:47-62. [PMID: 28583723 DOI: 10.1016/j.cytogfr.2017.04.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 04/07/2017] [Indexed: 12/21/2022]
Abstract
Mechanistic target of rapamycin (mTOR) is a serine/threonine kinase originally discovered as the molecular target of the immunosuppressant rapamycin. mTOR forms two compositionally and functionally distinct complexes, mTORC1 and mTORC2, which are crucial for coordinating nutrient, energy, oxygen, and growth factor availability with cellular growth, proliferation, and survival. Recent studies have identified critical, non-redundant roles for mTORC1 and mTORC2 in controlling B cell development, differentiation, and functions, and have highlighted emerging roles of the Folliculin-Fnip protein complex in regulating mTOR and B cell development. In this review, we summarize the basic mechanisms of mTOR signaling; describe what is known about the roles of mTORC1, mTORC2, and the Folliculin/Fnip1 pathway in B cell development and functions; and briefly outline current clinical approaches for targeting mTOR in B cell neoplasms. We conclude by highlighting a few salient questions and future perspectives regarding mTOR in B lineage cells.
Collapse
|
216
|
Aw A, Brown JR. The potential combination of BCL-2 inhibitors and ibrutinib as frontline therapy in chronic lymphocytic leukemia. Leuk Lymphoma 2017; 58:2287-2297. [PMID: 28482721 DOI: 10.1080/10428194.2017.1312387] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The recent development of small molecule inhibitors targeted at the B-cell receptor (BCR) pathway and the anti-apoptotic protein BCL-2 has revolutionized the care of patients with chronic lymphocytic leukemia (CLL). While durable responses to the BCR inhibitor ibrutinib have been observed in both previously untreated and relapsed/refractory CLL patients, residual disease is common in patients treated with single-agent ibrutinib. Interest remains high in therapeutic combinations that may lead to better quality remissions. A potential partner to ibrutinib with a distinct mechanism of action that is likely to lead to deeper responses is the BCL-2 inhibitor venetoclax. Preclinical studies have suggested synergism between inhibitors of BCR and BCL-2 and have paved the way to the development of ongoing clinical trials aimed at evaluating the combination of ibrutinib with venetoclax in CLL patients.
Collapse
Affiliation(s)
- Andrew Aw
- a Department of Medicine, Division of Hematology , The Ottawa Hospital, University of Ottawa , Ottawa , Canada
| | - Jennifer R Brown
- b CLL Center, Department of Medical Oncology , Dana-Farber Cancer Institute , Boston , MA , USA.,c Department of Medicine , Harvard Medical School , Boston , MA , USA
| |
Collapse
|
217
|
Lee S, Day NS, Miles RR, Perkins SL, Lim MS, Ayello J, van de Ven C, Harrison L, El-Mallawany NK, Goldman S, Cairo MS. Comparative genomic expression signatures of signal transduction pathways and targets in paediatric Burkitt lymphoma: a Children's Oncology Group report. Br J Haematol 2017; 177:601-611. [PMID: 28474336 DOI: 10.1111/bjh.14604] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 12/01/2016] [Indexed: 02/02/2023]
Abstract
Burkitt lymphoma (BL) is the most common histological subtype of non-Hodgkin lymphoma (NHL) in children and adolescents. Through the introduction of short intensive multi-agent chemoimmunotherapy, survival has improved significantly over the past 30 years. However, this successful approach is limited by significant chemotherapy-induced acute toxicity and risk of developing resistant disease, demonstrating the need to identify less toxic and targeted therapies. We analysed the comparative genomic signature and targetable signalling pathways in paediatric BL (PEBL) samples from the Children's Oncology Group study (ANHL01P1) by genomic profiling and selected genes were confirmed by quantitative real time polymerase chain reaction. These results were compared to PEBL samples from public databases and utilised the Gene Expression Omnibus (GEO) Series (GSE) 10172 and 4475 (n = 16), and 4732 (n = 15). Three hundred and seventy-six genes (approximately 25%) were similarly expressed among three PEBL sample groups. Several target genes in Toll-like receptor signalling, JAK-STAT signalling and MAPK signalling were significantly overexpressed in PEBL. In addition, several tyrosine kinases, including Bruton tyrosine kinase, protein tyrosine phosphatase and histone deacetylase inhibitor were highly expressed in PEBL. These pre-clinical results suggest that specific signal transduction pathways are overly expressed in PEBL and several pathways could serve as potential future therapeutic targets.
Collapse
Affiliation(s)
- Sanghoon Lee
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA.,Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
| | - Nancy S Day
- Department of Pediatrics, Columbia University, New York, NY, USA
| | - Rodney R Miles
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Sherrie L Perkins
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Megan S Lim
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Janet Ayello
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA
| | | | - Lauren Harrison
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA
| | | | | | - Mitchell S Cairo
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA.,Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA.,Departments of Medicine, Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| |
Collapse
|
218
|
Alfaro J, Pérez de Arce F, Belmar S, Fuentealba G, Avila P, Ureta G, Flores C, Acuña C, Delgado L, Gaete D, Pujala B, Barde A, Nayak AK, Upendra TVR, Patel D, Chauhan S, Sharma VK, Kanno S, Almirez RG, Hung DT, Chakravarty S, Rai R, Bernales S, Quinn KP, Pham SM, McCullagh E. Dual Inhibition of Bruton's Tyrosine Kinase and Phosphoinositide-3-Kinase p110 δ as a Therapeutic Approach to Treat Non-Hodgkin's B Cell Malignancies. J Pharmacol Exp Ther 2017; 361:312-321. [PMID: 28298527 DOI: 10.1124/jpet.116.238022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 03/10/2017] [Indexed: 02/01/2023] Open
Abstract
Although new targeted therapies, such as ibrutinib and idelalisib, have made a large impact on non-Hodgkin's lymphoma (NHL) patients, the disease is often fatal because patients are initially resistant to these targeted therapies, or because they eventually develop resistance. New drugs and treatments are necessary for these patients. One attractive approach is to inhibit multiple parallel pathways that drive the growth of these hematologic tumors, possibly prolonging the duration of the response and reducing resistance. Early clinical trials have tested this approach by dosing two drugs in combination in NHL patients. We discovered a single molecule, MDVN1003 (1-(5-amino-2,3-dihydro-1H-inden-2-yl)-3-(8-fluoro-3,4-dihydro-2H-benzo[b][1,4]oxazin-6-yl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine), that inhibits Bruton's tyrosine kinase and phosphatidylinositol-3-kinase δ, two proteins regulated by the B cell receptor that drive the growth of many NHLs. In this report, we show that this dual inhibitor prevents the activation of B cells and inhibits the phosphorylation of protein kinase B and extracellular signal-regulated kinase 1/2, two downstream mediators that are important for this process. Additionally, MDVN1003 induces cell death in a B cell lymphoma cell line but not in an irrelevant erythroblast cell line. Importantly, we found that this orally bioavailable dual inhibitor reduced tumor growth in a B cell lymphoma xenograft model more effectively than either ibrutinib or idelalisib. Taken together, these results suggest that dual inhibition of these two key pathways by a single molecule could be a viable approach for treatment of NHL patients.
Collapse
Affiliation(s)
- Jennifer Alfaro
- Translational Research Group, Fundación Ciencia y Vida, Santiago, Chile (J.A., F.P.d.A., S.Bel., G.F., P.A., G.U., C.F., C.A., L.D., D.G.); Biological Sciences Department, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Región de Valparaíso, Chile (F.P.d.A., S.Bel.); Chemistry Group, Integral BioSciences, Pvt. Ltd., Noida, India (B.P., A.B., A.K.N., T.V.R.U., D.P., S.C., V.K.S.); and Discovery Research, Medivation, Inc., now Pfizer, San Francisco, California (S.K., R.G.A., D.T.H., S.C., R.R., S.Ber., K.P.Q., S.M.P., E.M.)
| | - Felipe Pérez de Arce
- Translational Research Group, Fundación Ciencia y Vida, Santiago, Chile (J.A., F.P.d.A., S.Bel., G.F., P.A., G.U., C.F., C.A., L.D., D.G.); Biological Sciences Department, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Región de Valparaíso, Chile (F.P.d.A., S.Bel.); Chemistry Group, Integral BioSciences, Pvt. Ltd., Noida, India (B.P., A.B., A.K.N., T.V.R.U., D.P., S.C., V.K.S.); and Discovery Research, Medivation, Inc., now Pfizer, San Francisco, California (S.K., R.G.A., D.T.H., S.C., R.R., S.Ber., K.P.Q., S.M.P., E.M.)
| | - Sebastián Belmar
- Translational Research Group, Fundación Ciencia y Vida, Santiago, Chile (J.A., F.P.d.A., S.Bel., G.F., P.A., G.U., C.F., C.A., L.D., D.G.); Biological Sciences Department, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Región de Valparaíso, Chile (F.P.d.A., S.Bel.); Chemistry Group, Integral BioSciences, Pvt. Ltd., Noida, India (B.P., A.B., A.K.N., T.V.R.U., D.P., S.C., V.K.S.); and Discovery Research, Medivation, Inc., now Pfizer, San Francisco, California (S.K., R.G.A., D.T.H., S.C., R.R., S.Ber., K.P.Q., S.M.P., E.M.)
| | - Glenda Fuentealba
- Translational Research Group, Fundación Ciencia y Vida, Santiago, Chile (J.A., F.P.d.A., S.Bel., G.F., P.A., G.U., C.F., C.A., L.D., D.G.); Biological Sciences Department, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Región de Valparaíso, Chile (F.P.d.A., S.Bel.); Chemistry Group, Integral BioSciences, Pvt. Ltd., Noida, India (B.P., A.B., A.K.N., T.V.R.U., D.P., S.C., V.K.S.); and Discovery Research, Medivation, Inc., now Pfizer, San Francisco, California (S.K., R.G.A., D.T.H., S.C., R.R., S.Ber., K.P.Q., S.M.P., E.M.)
| | - Patricio Avila
- Translational Research Group, Fundación Ciencia y Vida, Santiago, Chile (J.A., F.P.d.A., S.Bel., G.F., P.A., G.U., C.F., C.A., L.D., D.G.); Biological Sciences Department, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Región de Valparaíso, Chile (F.P.d.A., S.Bel.); Chemistry Group, Integral BioSciences, Pvt. Ltd., Noida, India (B.P., A.B., A.K.N., T.V.R.U., D.P., S.C., V.K.S.); and Discovery Research, Medivation, Inc., now Pfizer, San Francisco, California (S.K., R.G.A., D.T.H., S.C., R.R., S.Ber., K.P.Q., S.M.P., E.M.)
| | - Gonzalo Ureta
- Translational Research Group, Fundación Ciencia y Vida, Santiago, Chile (J.A., F.P.d.A., S.Bel., G.F., P.A., G.U., C.F., C.A., L.D., D.G.); Biological Sciences Department, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Región de Valparaíso, Chile (F.P.d.A., S.Bel.); Chemistry Group, Integral BioSciences, Pvt. Ltd., Noida, India (B.P., A.B., A.K.N., T.V.R.U., D.P., S.C., V.K.S.); and Discovery Research, Medivation, Inc., now Pfizer, San Francisco, California (S.K., R.G.A., D.T.H., S.C., R.R., S.Ber., K.P.Q., S.M.P., E.M.)
| | - Camila Flores
- Translational Research Group, Fundación Ciencia y Vida, Santiago, Chile (J.A., F.P.d.A., S.Bel., G.F., P.A., G.U., C.F., C.A., L.D., D.G.); Biological Sciences Department, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Región de Valparaíso, Chile (F.P.d.A., S.Bel.); Chemistry Group, Integral BioSciences, Pvt. Ltd., Noida, India (B.P., A.B., A.K.N., T.V.R.U., D.P., S.C., V.K.S.); and Discovery Research, Medivation, Inc., now Pfizer, San Francisco, California (S.K., R.G.A., D.T.H., S.C., R.R., S.Ber., K.P.Q., S.M.P., E.M.)
| | - Claudia Acuña
- Translational Research Group, Fundación Ciencia y Vida, Santiago, Chile (J.A., F.P.d.A., S.Bel., G.F., P.A., G.U., C.F., C.A., L.D., D.G.); Biological Sciences Department, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Región de Valparaíso, Chile (F.P.d.A., S.Bel.); Chemistry Group, Integral BioSciences, Pvt. Ltd., Noida, India (B.P., A.B., A.K.N., T.V.R.U., D.P., S.C., V.K.S.); and Discovery Research, Medivation, Inc., now Pfizer, San Francisco, California (S.K., R.G.A., D.T.H., S.C., R.R., S.Ber., K.P.Q., S.M.P., E.M.)
| | - Luz Delgado
- Translational Research Group, Fundación Ciencia y Vida, Santiago, Chile (J.A., F.P.d.A., S.Bel., G.F., P.A., G.U., C.F., C.A., L.D., D.G.); Biological Sciences Department, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Región de Valparaíso, Chile (F.P.d.A., S.Bel.); Chemistry Group, Integral BioSciences, Pvt. Ltd., Noida, India (B.P., A.B., A.K.N., T.V.R.U., D.P., S.C., V.K.S.); and Discovery Research, Medivation, Inc., now Pfizer, San Francisco, California (S.K., R.G.A., D.T.H., S.C., R.R., S.Ber., K.P.Q., S.M.P., E.M.)
| | - Diana Gaete
- Translational Research Group, Fundación Ciencia y Vida, Santiago, Chile (J.A., F.P.d.A., S.Bel., G.F., P.A., G.U., C.F., C.A., L.D., D.G.); Biological Sciences Department, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Región de Valparaíso, Chile (F.P.d.A., S.Bel.); Chemistry Group, Integral BioSciences, Pvt. Ltd., Noida, India (B.P., A.B., A.K.N., T.V.R.U., D.P., S.C., V.K.S.); and Discovery Research, Medivation, Inc., now Pfizer, San Francisco, California (S.K., R.G.A., D.T.H., S.C., R.R., S.Ber., K.P.Q., S.M.P., E.M.)
| | - Brahmam Pujala
- Translational Research Group, Fundación Ciencia y Vida, Santiago, Chile (J.A., F.P.d.A., S.Bel., G.F., P.A., G.U., C.F., C.A., L.D., D.G.); Biological Sciences Department, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Región de Valparaíso, Chile (F.P.d.A., S.Bel.); Chemistry Group, Integral BioSciences, Pvt. Ltd., Noida, India (B.P., A.B., A.K.N., T.V.R.U., D.P., S.C., V.K.S.); and Discovery Research, Medivation, Inc., now Pfizer, San Francisco, California (S.K., R.G.A., D.T.H., S.C., R.R., S.Ber., K.P.Q., S.M.P., E.M.)
| | - Anup Barde
- Translational Research Group, Fundación Ciencia y Vida, Santiago, Chile (J.A., F.P.d.A., S.Bel., G.F., P.A., G.U., C.F., C.A., L.D., D.G.); Biological Sciences Department, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Región de Valparaíso, Chile (F.P.d.A., S.Bel.); Chemistry Group, Integral BioSciences, Pvt. Ltd., Noida, India (B.P., A.B., A.K.N., T.V.R.U., D.P., S.C., V.K.S.); and Discovery Research, Medivation, Inc., now Pfizer, San Francisco, California (S.K., R.G.A., D.T.H., S.C., R.R., S.Ber., K.P.Q., S.M.P., E.M.)
| | - Anjan K Nayak
- Translational Research Group, Fundación Ciencia y Vida, Santiago, Chile (J.A., F.P.d.A., S.Bel., G.F., P.A., G.U., C.F., C.A., L.D., D.G.); Biological Sciences Department, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Región de Valparaíso, Chile (F.P.d.A., S.Bel.); Chemistry Group, Integral BioSciences, Pvt. Ltd., Noida, India (B.P., A.B., A.K.N., T.V.R.U., D.P., S.C., V.K.S.); and Discovery Research, Medivation, Inc., now Pfizer, San Francisco, California (S.K., R.G.A., D.T.H., S.C., R.R., S.Ber., K.P.Q., S.M.P., E.M.)
| | - T V R Upendra
- Translational Research Group, Fundación Ciencia y Vida, Santiago, Chile (J.A., F.P.d.A., S.Bel., G.F., P.A., G.U., C.F., C.A., L.D., D.G.); Biological Sciences Department, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Región de Valparaíso, Chile (F.P.d.A., S.Bel.); Chemistry Group, Integral BioSciences, Pvt. Ltd., Noida, India (B.P., A.B., A.K.N., T.V.R.U., D.P., S.C., V.K.S.); and Discovery Research, Medivation, Inc., now Pfizer, San Francisco, California (S.K., R.G.A., D.T.H., S.C., R.R., S.Ber., K.P.Q., S.M.P., E.M.)
| | - Dhananjay Patel
- Translational Research Group, Fundación Ciencia y Vida, Santiago, Chile (J.A., F.P.d.A., S.Bel., G.F., P.A., G.U., C.F., C.A., L.D., D.G.); Biological Sciences Department, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Región de Valparaíso, Chile (F.P.d.A., S.Bel.); Chemistry Group, Integral BioSciences, Pvt. Ltd., Noida, India (B.P., A.B., A.K.N., T.V.R.U., D.P., S.C., V.K.S.); and Discovery Research, Medivation, Inc., now Pfizer, San Francisco, California (S.K., R.G.A., D.T.H., S.C., R.R., S.Ber., K.P.Q., S.M.P., E.M.)
| | - Shailender Chauhan
- Translational Research Group, Fundación Ciencia y Vida, Santiago, Chile (J.A., F.P.d.A., S.Bel., G.F., P.A., G.U., C.F., C.A., L.D., D.G.); Biological Sciences Department, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Región de Valparaíso, Chile (F.P.d.A., S.Bel.); Chemistry Group, Integral BioSciences, Pvt. Ltd., Noida, India (B.P., A.B., A.K.N., T.V.R.U., D.P., S.C., V.K.S.); and Discovery Research, Medivation, Inc., now Pfizer, San Francisco, California (S.K., R.G.A., D.T.H., S.C., R.R., S.Ber., K.P.Q., S.M.P., E.M.)
| | - Vijay K Sharma
- Translational Research Group, Fundación Ciencia y Vida, Santiago, Chile (J.A., F.P.d.A., S.Bel., G.F., P.A., G.U., C.F., C.A., L.D., D.G.); Biological Sciences Department, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Región de Valparaíso, Chile (F.P.d.A., S.Bel.); Chemistry Group, Integral BioSciences, Pvt. Ltd., Noida, India (B.P., A.B., A.K.N., T.V.R.U., D.P., S.C., V.K.S.); and Discovery Research, Medivation, Inc., now Pfizer, San Francisco, California (S.K., R.G.A., D.T.H., S.C., R.R., S.Ber., K.P.Q., S.M.P., E.M.)
| | - Stacy Kanno
- Translational Research Group, Fundación Ciencia y Vida, Santiago, Chile (J.A., F.P.d.A., S.Bel., G.F., P.A., G.U., C.F., C.A., L.D., D.G.); Biological Sciences Department, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Región de Valparaíso, Chile (F.P.d.A., S.Bel.); Chemistry Group, Integral BioSciences, Pvt. Ltd., Noida, India (B.P., A.B., A.K.N., T.V.R.U., D.P., S.C., V.K.S.); and Discovery Research, Medivation, Inc., now Pfizer, San Francisco, California (S.K., R.G.A., D.T.H., S.C., R.R., S.Ber., K.P.Q., S.M.P., E.M.)
| | - Ramona G Almirez
- Translational Research Group, Fundación Ciencia y Vida, Santiago, Chile (J.A., F.P.d.A., S.Bel., G.F., P.A., G.U., C.F., C.A., L.D., D.G.); Biological Sciences Department, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Región de Valparaíso, Chile (F.P.d.A., S.Bel.); Chemistry Group, Integral BioSciences, Pvt. Ltd., Noida, India (B.P., A.B., A.K.N., T.V.R.U., D.P., S.C., V.K.S.); and Discovery Research, Medivation, Inc., now Pfizer, San Francisco, California (S.K., R.G.A., D.T.H., S.C., R.R., S.Ber., K.P.Q., S.M.P., E.M.)
| | - David T Hung
- Translational Research Group, Fundación Ciencia y Vida, Santiago, Chile (J.A., F.P.d.A., S.Bel., G.F., P.A., G.U., C.F., C.A., L.D., D.G.); Biological Sciences Department, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Región de Valparaíso, Chile (F.P.d.A., S.Bel.); Chemistry Group, Integral BioSciences, Pvt. Ltd., Noida, India (B.P., A.B., A.K.N., T.V.R.U., D.P., S.C., V.K.S.); and Discovery Research, Medivation, Inc., now Pfizer, San Francisco, California (S.K., R.G.A., D.T.H., S.C., R.R., S.Ber., K.P.Q., S.M.P., E.M.)
| | - Sarvajit Chakravarty
- Translational Research Group, Fundación Ciencia y Vida, Santiago, Chile (J.A., F.P.d.A., S.Bel., G.F., P.A., G.U., C.F., C.A., L.D., D.G.); Biological Sciences Department, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Región de Valparaíso, Chile (F.P.d.A., S.Bel.); Chemistry Group, Integral BioSciences, Pvt. Ltd., Noida, India (B.P., A.B., A.K.N., T.V.R.U., D.P., S.C., V.K.S.); and Discovery Research, Medivation, Inc., now Pfizer, San Francisco, California (S.K., R.G.A., D.T.H., S.C., R.R., S.Ber., K.P.Q., S.M.P., E.M.)
| | - Roopa Rai
- Translational Research Group, Fundación Ciencia y Vida, Santiago, Chile (J.A., F.P.d.A., S.Bel., G.F., P.A., G.U., C.F., C.A., L.D., D.G.); Biological Sciences Department, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Región de Valparaíso, Chile (F.P.d.A., S.Bel.); Chemistry Group, Integral BioSciences, Pvt. Ltd., Noida, India (B.P., A.B., A.K.N., T.V.R.U., D.P., S.C., V.K.S.); and Discovery Research, Medivation, Inc., now Pfizer, San Francisco, California (S.K., R.G.A., D.T.H., S.C., R.R., S.Ber., K.P.Q., S.M.P., E.M.)
| | - Sebastián Bernales
- Translational Research Group, Fundación Ciencia y Vida, Santiago, Chile (J.A., F.P.d.A., S.Bel., G.F., P.A., G.U., C.F., C.A., L.D., D.G.); Biological Sciences Department, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Región de Valparaíso, Chile (F.P.d.A., S.Bel.); Chemistry Group, Integral BioSciences, Pvt. Ltd., Noida, India (B.P., A.B., A.K.N., T.V.R.U., D.P., S.C., V.K.S.); and Discovery Research, Medivation, Inc., now Pfizer, San Francisco, California (S.K., R.G.A., D.T.H., S.C., R.R., S.Ber., K.P.Q., S.M.P., E.M.)
| | - Kevin P Quinn
- Translational Research Group, Fundación Ciencia y Vida, Santiago, Chile (J.A., F.P.d.A., S.Bel., G.F., P.A., G.U., C.F., C.A., L.D., D.G.); Biological Sciences Department, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Región de Valparaíso, Chile (F.P.d.A., S.Bel.); Chemistry Group, Integral BioSciences, Pvt. Ltd., Noida, India (B.P., A.B., A.K.N., T.V.R.U., D.P., S.C., V.K.S.); and Discovery Research, Medivation, Inc., now Pfizer, San Francisco, California (S.K., R.G.A., D.T.H., S.C., R.R., S.Ber., K.P.Q., S.M.P., E.M.)
| | - Son M Pham
- Translational Research Group, Fundación Ciencia y Vida, Santiago, Chile (J.A., F.P.d.A., S.Bel., G.F., P.A., G.U., C.F., C.A., L.D., D.G.); Biological Sciences Department, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Región de Valparaíso, Chile (F.P.d.A., S.Bel.); Chemistry Group, Integral BioSciences, Pvt. Ltd., Noida, India (B.P., A.B., A.K.N., T.V.R.U., D.P., S.C., V.K.S.); and Discovery Research, Medivation, Inc., now Pfizer, San Francisco, California (S.K., R.G.A., D.T.H., S.C., R.R., S.Ber., K.P.Q., S.M.P., E.M.)
| | - Emma McCullagh
- Translational Research Group, Fundación Ciencia y Vida, Santiago, Chile (J.A., F.P.d.A., S.Bel., G.F., P.A., G.U., C.F., C.A., L.D., D.G.); Biological Sciences Department, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Región de Valparaíso, Chile (F.P.d.A., S.Bel.); Chemistry Group, Integral BioSciences, Pvt. Ltd., Noida, India (B.P., A.B., A.K.N., T.V.R.U., D.P., S.C., V.K.S.); and Discovery Research, Medivation, Inc., now Pfizer, San Francisco, California (S.K., R.G.A., D.T.H., S.C., R.R., S.Ber., K.P.Q., S.M.P., E.M.)
| |
Collapse
|
219
|
Kuo HP, Ezell SA, Schweighofer KJ, Cheung LWK, Hsieh S, Apatira M, Sirisawad M, Eckert K, Hsu SJ, Chen CT, Beaupre DM, Versele M, Chang BY. Combination of Ibrutinib and ABT-199 in Diffuse Large B-Cell Lymphoma and Follicular Lymphoma. Mol Cancer Ther 2017; 16:1246-1256. [PMID: 28428442 DOI: 10.1158/1535-7163.mct-16-0555] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 02/01/2017] [Accepted: 04/14/2017] [Indexed: 11/16/2022]
Abstract
Diffuse large B-cell lymphoma (DLBCL) and follicular lymphoma are the most prevalent B-lymphocyte neoplasms in which abnormal activation of the Bruton tyrosine kinase (BTK)-mediated B-cell receptor signaling pathway contributes to pathogenesis. Ibrutinib is an oral covalent BTK inhibitor that has shown some efficacy in both indications. To improve ibrutinib efficacy through combination therapy, we first investigated differential gene expression in parental and ibrutinib-resistant cell lines to better understand the mechanisms of resistance. Ibrutinib-resistant TMD8 cells had higher BCL2 gene expression and increased sensitivity to ABT-199, a BCL-2 inhibitor. Consistently, clinical samples from ABC-DLBCL patients who experienced poorer response to ibrutinib had higher BCL2 gene expression. We further demonstrated synergistic growth suppression by ibrutinib and ABT-199 in multiple ABC-DLBCL, GCB-DLBCL, and follicular lymphoma cell lines. The combination of both drugs also reduced colony formation, increased apoptosis, and inhibited tumor growth in a TMD8 xenograft model. A synergistic combination effect was also found in ibrutinib-resistant cells generated by either genetic mutation or drug treatment. Together, these findings suggest a potential clinical benefit from ibrutinib and ABT-199 combination therapy. Mol Cancer Ther; 16(7); 1246-56. ©2017 AACR.
Collapse
Affiliation(s)
- Hsu-Ping Kuo
- Research Department, Pharmacyclics LLC, an AbbVie Company, Sunnyvale, California
| | - Scott A Ezell
- Research Department, Pharmacyclics LLC, an AbbVie Company, Sunnyvale, California
| | - Karl J Schweighofer
- Research Department, Pharmacyclics LLC, an AbbVie Company, Sunnyvale, California
| | - Leo W K Cheung
- Research Department, Pharmacyclics LLC, an AbbVie Company, Sunnyvale, California
| | - Sidney Hsieh
- Research Department, Pharmacyclics LLC, an AbbVie Company, Sunnyvale, California
| | - Mutiah Apatira
- Research Department, Pharmacyclics LLC, an AbbVie Company, Sunnyvale, California
| | - Mint Sirisawad
- Research Department, Pharmacyclics LLC, an AbbVie Company, Sunnyvale, California
| | - Karl Eckert
- Research Department, Pharmacyclics LLC, an AbbVie Company, Sunnyvale, California
| | - Ssucheng J Hsu
- Research Department, Pharmacyclics LLC, an AbbVie Company, Sunnyvale, California
| | - Chun-Te Chen
- Research Department, Pharmacyclics LLC, an AbbVie Company, Sunnyvale, California
| | - Darrin M Beaupre
- Research Department, Pharmacyclics LLC, an AbbVie Company, Sunnyvale, California
| | | | - Betty Y Chang
- Research Department, Pharmacyclics LLC, an AbbVie Company, Sunnyvale, California.
| |
Collapse
|
220
|
Nelson EA, Dyall J, Hoenen T, Barnes AB, Zhou H, Liang JY, Michelotti J, Dewey WH, DeWald LE, Bennett RS, Morris PJ, Guha R, Klumpp-Thomas C, McKnight C, Chen YC, Xu X, Wang A, Hughes E, Martin S, Thomas C, Jahrling PB, Hensley LE, Olinger GG, White JM. The phosphatidylinositol-3-phosphate 5-kinase inhibitor apilimod blocks filoviral entry and infection. PLoS Negl Trop Dis 2017; 11:e0005540. [PMID: 28403145 PMCID: PMC5402990 DOI: 10.1371/journal.pntd.0005540] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 04/24/2017] [Accepted: 03/30/2017] [Indexed: 12/12/2022] Open
Abstract
Phosphatidylinositol-3-phosphate 5-kinase (PIKfyve) is a lipid kinase involved in endosome maturation that emerged from a haploid genetic screen as being required for Ebola virus (EBOV) infection. Here we analyzed the effects of apilimod, a PIKfyve inhibitor that was reported to be well tolerated in humans in phase 2 clinical trials, for its effects on entry and infection of EBOV and Marburg virus (MARV). We first found that apilimod blocks infections by EBOV and MARV in Huh 7, Vero E6 and primary human macrophage cells, with notable potency in the macrophages (IC50, 10 nM). We next observed that similar doses of apilimod block EBOV-glycoprotein-virus like particle (VLP) entry and transcription-replication competent VLP infection, suggesting that the primary mode of action of apilimod is as an entry inhibitor, preventing release of the viral genome into the cytoplasm to initiate replication. After providing evidence that the anti-EBOV action of apilimod is via PIKfyve, we showed that it blocks trafficking of EBOV VLPs to endolysosomes containing Niemann-Pick C1 (NPC1), the intracellular receptor for EBOV. Concurrently apilimod caused VLPs to accumulate in early endosome antigen 1-positive endosomes. We did not detect any effects of apilimod on bulk endosome acidification, on the activity of cathepsins B and L, or on cholesterol export from endolysosomes. Hence by antagonizing PIKfyve, apilimod appears to block EBOV trafficking to its site of fusion and entry into the cytoplasm. Given the drug's observed anti-filoviral activity, relatively unexplored mechanism of entry inhibition, and reported tolerability in humans, we propose that apilimod be further explored as part of a therapeutic regimen to treat filoviral infections.
Collapse
Affiliation(s)
- Elizabeth A. Nelson
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Julie Dyall
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Thomas Hoenen
- Laboratory of Virology, Division of Intramural Research, National Institutes of Health, Hamilton, Montana, United States of America
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald–Insel Riems, Germany
| | - Alyson B. Barnes
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Huanying Zhou
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Janie Y. Liang
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Julia Michelotti
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - William H. Dewey
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Lisa Evans DeWald
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Richard S. Bennett
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Patrick J. Morris
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Rajarshi Guha
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Carleen Klumpp-Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Crystal McKnight
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yu-Chi Chen
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Xin Xu
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Amy Wang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Emma Hughes
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Scott Martin
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Craig Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Peter B. Jahrling
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Lisa E. Hensley
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Gene G. Olinger
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Judith M. White
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, United States of America
| |
Collapse
|
221
|
Wu Z, Zhao S, Fash DM, Li Z, Chain WJ, Beutler JA. Englerins: A Comprehensive Review. JOURNAL OF NATURAL PRODUCTS 2017; 80:771-781. [PMID: 28170253 PMCID: PMC6198806 DOI: 10.1021/acs.jnatprod.6b01167] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
In the decade since the discovery of englerin A (1) and its potent activity in cancer models, this natural product and its analogues have been the subject of numerous chemical, biological, and preclinical studies by many research groups. This review summarizes published findings and proposes further research directions required for entry of an englerin analogue into clinical trials for kidney cancer and other conditions.
Collapse
Affiliation(s)
- Zhenhua Wu
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, United States
| | - Senzhi Zhao
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, United States
| | - David M. Fash
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, United States
| | - Zhenwu Li
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, United States
| | - William J. Chain
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, United States
| | - John A. Beutler
- Molecular Targets Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| |
Collapse
|
222
|
de Bruijn MJW, Rip J, van der Ploeg EK, van Greuningen LW, Ta VTB, Kil LP, Langerak AW, Rimmelzwaan GF, Ellmeier W, Hendriks RW, Corneth OBJ. Distinct and Overlapping Functions of TEC Kinase and BTK in B Cell Receptor Signaling. THE JOURNAL OF IMMUNOLOGY 2017; 198:3058-3068. [PMID: 28275136 DOI: 10.4049/jimmunol.1601285] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 02/15/2017] [Indexed: 01/18/2023]
Abstract
The Tec tyrosine kinase is expressed in many cell types, including hematopoietic cells, and is a member of the Tec kinase family that also includes Btk. Although the role of Btk in B cells has been extensively studied, the role of Tec kinase in B cells remains largely unclear. It was previously shown that Tec kinase has the ability to partly compensate for loss of Btk activity in B cell differentiation, although the underlying mechanism is unknown. In this study, we confirm that Tec kinase is not essential for normal B cell development when Btk is present, but we also found that Tec-deficient mature B cells showed increased activation, proliferation, and survival upon BCR stimulation, even in the presence of Btk. Whereas Tec deficiency did not affect phosphorylation of phospholipase Cγ or Ca2+ influx, it was associated with significantly increased activation of the intracellular Akt/S6 kinase signaling pathway upon BCR and CD40 stimulation. The increased S6 kinase phosphorylation in Tec-deficient B cells was dependent on Btk kinase activity, as ibrutinib treatment restored pS6 to wild-type levels, although Btk protein and phosphorylation levels were comparable to controls. In Tec-deficient mice in vivo, B cell responses to model Ags and humoral immunity upon influenza infection were enhanced. Moreover, aged mice lacking Tec kinase developed a mild autoimmune phenotype. Taken together, these data indicate that in mature B cells, Tec and Btk may compete for activation of the Akt signaling pathway, whereby the activating capacity of Btk is limited by the presence of Tec kinase.
Collapse
Affiliation(s)
- Marjolein J W de Bruijn
- Department of Pulmonary Medicine, Erasmus University Medical Center, 3015 GE Rotterdam, the Netherlands
| | - Jasper Rip
- Department of Pulmonary Medicine, Erasmus University Medical Center, 3015 GE Rotterdam, the Netherlands
| | - Esmee K van der Ploeg
- Department of Pulmonary Medicine, Erasmus University Medical Center, 3015 GE Rotterdam, the Netherlands
| | - Lars W van Greuningen
- Department of Pulmonary Medicine, Erasmus University Medical Center, 3015 GE Rotterdam, the Netherlands
| | - Van T B Ta
- Department of Pulmonary Medicine, Erasmus University Medical Center, 3015 GE Rotterdam, the Netherlands
| | - Laurens P Kil
- Department of Pulmonary Medicine, Erasmus University Medical Center, 3015 GE Rotterdam, the Netherlands
| | - Anton W Langerak
- Department of Immunology, Erasmus University Medical Center, 3015 GE Rotterdam, the Netherlands
| | - Guus F Rimmelzwaan
- Department of Viroscience, Erasmus University Medical Center, 3000 CA Rotterdam, the Netherlands; and
| | - Wilfried Ellmeier
- Division of Immunobiology, Institute of Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus University Medical Center, 3015 GE Rotterdam, the Netherlands;
| | - Odilia B J Corneth
- Department of Pulmonary Medicine, Erasmus University Medical Center, 3015 GE Rotterdam, the Netherlands
| |
Collapse
|
223
|
Karmali R, Gordon LI. Molecular Subtyping in Diffuse Large B Cell Lymphoma: Closer to an Approach of Precision Therapy. Curr Treat Options Oncol 2017; 18:11. [DOI: 10.1007/s11864-017-0449-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
224
|
Lal-Nag M, McGee L, Guha R, Lengyel E, Kenny HA, Ferrer M. A High-Throughput Screening Model of the Tumor Microenvironment for Ovarian Cancer Cell Growth. SLAS DISCOVERY 2017; 22:494-506. [PMID: 28346091 DOI: 10.1177/2472555216687082] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The tumor microenvironment plays an important role in the processes of tumor growth, metastasis, and drug resistance. We have used a multilayered 3D primary cell culture model that reproduces the human ovarian cancer metastatic microenvironment to study the effect of the microenvironment on the pharmacological responses of different classes of drugs on cancer cell proliferation. A collection of oncology drugs was screened to identify compounds that inhibited the proliferation of ovarian cancer cells growing as monolayers or forming spheroids, on plastic and on a 3D microenvironment culture model of the omentum metastatic site, and also cells already in preformed spheroids. Target-based analysis of the pharmacological responses revealed that several classes of targets were more efficacious in cancer cells growing in the absence of the metastatic microenvironment, and other target classes were less efficacious in cancer cells in preformed spheres compared to forming spheroid cultures. These findings show that both the cellular context of the tumor microenvironment and cell adhesion mode have an essential role in cancer cell drug resistance. Therefore, it is important to perform screens for new drugs using model systems that more faithfully recapitulate the tissue composition at the site of tumor growth and metastasis.
Collapse
Affiliation(s)
- Madhu Lal-Nag
- 1 Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, Bethesda, MD, USA
| | - Lauren McGee
- 1 Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, Bethesda, MD, USA
| | - Rajarshi Guha
- 1 Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, Bethesda, MD, USA
| | - Ernst Lengyel
- 2 Department of Obstetrics and Gynecology/Section of Gynecologic Oncology, University of Chicago, Chicago, IL, USA
| | - Hilary A Kenny
- 2 Department of Obstetrics and Gynecology/Section of Gynecologic Oncology, University of Chicago, Chicago, IL, USA
| | - Marc Ferrer
- 1 Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
225
|
Du W, Goldstein R, Jiang Y, Aly O, Cerchietti L, Melnick A, Elemento O. Effective Combination Therapies for B-cell Lymphoma Predicted by a Virtual Disease Model. Cancer Res 2017; 77:1818-1830. [PMID: 28130226 DOI: 10.1158/0008-5472.can-16-0476] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 12/10/2016] [Accepted: 01/23/2017] [Indexed: 12/15/2022]
Abstract
The complexity of cancer signaling networks limits the efficacy of most single-agent treatments and brings about challenges in identifying effective combinatorial therapies. In this study, we used chronic active B-cell receptor (BCR) signaling in diffuse large B-cell lymphoma as a model system to establish a computational framework to optimize combinatorial therapy in silico We constructed a detailed kinetic model of the BCR signaling network, which captured the known complex cross-talk between the NFκB, ERK, and AKT pathways and multiple feedback loops. Combining this signaling model with a data-derived tumor growth model, we predicted viability responses of many single drug and drug combinations in agreement with experimental data. Under this framework, we exhaustively predicted and ranked the efficacy and synergism of all possible combinatorial inhibitions of eleven currently targetable kinases in the BCR signaling network. Ultimately, our work establishes a detailed kinetic model of the core BCR signaling network and provides the means to explore the large space of possible drug combinations. Cancer Res; 77(8); 1818-30. ©2017 AACR.
Collapse
Affiliation(s)
- Wei Du
- Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York
| | - Rebecca Goldstein
- Hematology/Oncology Division, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Yanwen Jiang
- Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York.,Hematology/Oncology Division, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Omar Aly
- Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York
| | - Leandro Cerchietti
- Hematology/Oncology Division, Department of Medicine, Weill Cornell Medicine, New York, New York.,Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York
| | - Ari Melnick
- Hematology/Oncology Division, Department of Medicine, Weill Cornell Medicine, New York, New York.,Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York
| | - Olivier Elemento
- Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York. .,Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York.,Institute for Precision Medicine, Weill Cornell Medicine, New York, New York
| |
Collapse
|
226
|
Abstract
The allure of phenotypic screening, combined with the industry preference for target-based approaches, has prompted the development of innovative chemical biology technologies that facilitate the identification of new therapeutic targets for accelerated drug discovery. A chemogenomic library is a collection of selective small-molecule pharmacological agents, and a hit from such a set in a phenotypic screen suggests that the annotated target or targets of that pharmacological agent may be involved in perturbing the observable phenotype. In this Review, we describe opportunities for chemogenomic screening to considerably expedite the conversion of phenotypic screening projects into target-based drug discovery approaches. Other applications are explored, including drug repositioning, predictive toxicology and the discovery of novel pharmacological modalities.
Collapse
|
227
|
Paul J, Soujon M, Wengner AM, Zitzmann-Kolbe S, Sturz A, Haike K, Keng Magdalene KH, Tan SH, Lange M, Tan SY, Mumberg D, Lim ST, Ziegelbauer K, Liu N. Simultaneous Inhibition of PI3Kδ and PI3Kα Induces ABC-DLBCL Regression by Blocking BCR-Dependent and -Independent Activation of NF-κB and AKT. Cancer Cell 2017; 31:64-78. [PMID: 28073005 DOI: 10.1016/j.ccell.2016.12.003] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 10/07/2016] [Accepted: 12/05/2016] [Indexed: 01/28/2023]
Abstract
Compared with follicular lymphoma, high PI3Kα expression was more prevalent in diffuse large B cell lymphoma (DLBCL), although both tumor types expressed substantial PI3Kδ. Simultaneous inhibition of PI3Kα and PI3Kδ dramatically enhanced the anti-tumor profile in ABC-DLBCL models compared with selective inhibition of PI3Kδ, PI3Kα, or BTK. The anti-tumor activity was associated with suppression of p-AKT and a mechanism of blocking nuclear factor-κB activation driven by CD79mut, CARD11mut, TNFAIP3mut, or MYD88mut. Inhibition of PI3Kα/δ resulted in tumor regression in an ibrutinib-resistant CD79BWT/MYD88mut patient-derived ABC-DLBCL model. Furthermore, rebound activation of BTK and AKT was identified as a mechanism limiting CD79Bmut-ABC-DLBCL to show a robust response to PI3K and BTK inhibitor monotherapies. A combination of ibrutinib with the PI3Kα/δ inhibitor copanlisib produced a sustained complete response in vivo in CD79Bmut/MYD88mut ABC-DLBCL models.
Collapse
MESH Headings
- Adenine/analogs & derivatives
- Adult
- Agammaglobulinaemia Tyrosine Kinase
- Aged
- Animals
- Cell Line, Tumor
- Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Humans
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/pathology
- Mice
- Mice, Inbred BALB C
- Middle Aged
- NF-kappa B/physiology
- Phosphoinositide-3 Kinase Inhibitors
- Piperidines
- Protein-Tyrosine Kinases/antagonists & inhibitors
- Proto-Oncogene Proteins c-akt/physiology
- Pyrazoles/pharmacology
- Pyrimidines/pharmacology
- Quinazolines/pharmacology
- Receptors, Antigen, B-Cell/physiology
Collapse
Affiliation(s)
- Juliane Paul
- Bayer AG, Drug Discovery Oncology, Muellerstrasse 178, 13353 Berlin, Germany
| | - Maurice Soujon
- Bayer AG, Drug Discovery Oncology, Muellerstrasse 178, 13353 Berlin, Germany
| | - Antje M Wengner
- Bayer AG, Drug Discovery Oncology, Muellerstrasse 178, 13353 Berlin, Germany
| | | | - Andrea Sturz
- Bayer AG, Drug Discovery Oncology, Muellerstrasse 178, 13353 Berlin, Germany
| | - Katja Haike
- Bayer AG, Drug Discovery Oncology, Muellerstrasse 178, 13353 Berlin, Germany
| | - Koh Hui Keng Magdalene
- Advanced Molecular Pathology Laboratory, Singapore Health Services Pte Ltd, 20 College Road, 169856 Singapore, Singapore
| | - Sze Huey Tan
- Clinical Trials and Epidemiological Sciences, National Cancer Centre Singapore, 11 Hospital Drive, 169610 Singapore, Singapore
| | - Martin Lange
- Bayer AG, Drug Discovery Oncology, Muellerstrasse 178, 13353 Berlin, Germany
| | - Soo Yong Tan
- Advanced Molecular Pathology Laboratory, Singapore Health Services Pte Ltd, 20 College Road, 169856 Singapore, Singapore
| | - Dominik Mumberg
- Bayer AG, Drug Discovery Oncology, Muellerstrasse 178, 13353 Berlin, Germany
| | - Soon Thye Lim
- Office of Education, Duke-NUS Graduate Medical School, 8 College Road, 169857 Singapore, Singapore
| | - Karl Ziegelbauer
- Bayer AG, Drug Discovery Oncology, Muellerstrasse 178, 13353 Berlin, Germany
| | - Ningshu Liu
- Bayer AG, Drug Discovery Oncology, Muellerstrasse 178, 13353 Berlin, Germany.
| |
Collapse
|
228
|
Tang J. Informatics Approaches for Predicting, Understanding, and Testing Cancer Drug Combinations. Methods Mol Biol 2017; 1636:485-506. [PMID: 28730498 DOI: 10.1007/978-1-4939-7154-1_30] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Making cancer treatment more effective is one of the grand challenges in our health care system. However, many drugs have entered clinical trials but so far showed limited efficacy or induced rapid development of resistance. We urgently need multi-targeted drug combinations, which shall selectively inhibit the cancer cells and block the emergence of drug resistance. The book chapter focuses on mathematical and computational tools to facilitate the discovery of the most promising drug combinations to improve efficacy and prevent resistance. Data integration approaches that leverage drug-target interactions, cancer molecular features, and signaling pathways for predicting, understanding, and testing drug combinations are critically reviewed.
Collapse
Affiliation(s)
- Jing Tang
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Tukholmankatu 8, 00290, Helsinki, Finland. .,Department of Mathematics and Statistics, University of Turku, Turku, Finland.
| |
Collapse
|
229
|
Ezell SA, Wang S, Bihani T, Lai Z, Grosskurth SE, Tepsuporn S, Davies BR, Huszar D, Byth KF. Differential regulation of mTOR signaling determines sensitivity to AKT inhibition in diffuse large B cell lymphoma. Oncotarget 2016; 7:9163-74. [PMID: 26824321 PMCID: PMC4891033 DOI: 10.18632/oncotarget.7036] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 01/19/2016] [Indexed: 12/04/2022] Open
Abstract
Agents that target components of the PI3K/AKT/mTOR pathway are under investigation for the treatment of diffuse large B cell lymphoma (DLBCL). Given the highly heterogeneous nature of DLBCL, it is not clear whether all subtypes of DLBCL will be susceptible to PI3K pathway inhibition, or which kinase within this pathway is the most favorable target. Pharmacological profiling of a panel of DLBCL cell lines revealed a subset of DLBCL that was resistant to AKT inhibition. Strikingly, sensitivity to AKT inhibitors correlated with the ability of these inhibitors to block phosphorylation of S6K1 and ribosomal protein S6. Cell lines resistant to AKT inhibition activated S6K1 independent of AKT either through upregulation of PIM2 or through activation by B cell receptor (BCR) signaling components. Finally, combined inhibition of AKT and BTK, PIM2, or S6K1 proved to be an effective strategy to overcome resistance to AKT inhibition in DLBCL.
Collapse
Affiliation(s)
- Scott A Ezell
- AstraZeneca Oncology, Waltham, Massachusetts, MA, USA
| | - Suping Wang
- AstraZeneca Oncology, Waltham, Massachusetts, MA, USA
| | - Teeru Bihani
- AstraZeneca Oncology, Waltham, Massachusetts, MA, USA
| | - Zhongwu Lai
- AstraZeneca Oncology, Waltham, Massachusetts, MA, USA
| | | | | | | | - Dennis Huszar
- AstraZeneca Oncology, Waltham, Massachusetts, MA, USA
| | - Kate F Byth
- AstraZeneca Oncology, Waltham, Massachusetts, MA, USA
| |
Collapse
|
230
|
Large-scale pharmacological profiling of 3D tumor models of cancer cells. Cell Death Dis 2016; 7:e2492. [PMID: 27906188 PMCID: PMC5261027 DOI: 10.1038/cddis.2016.360] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 08/22/2016] [Accepted: 09/19/2016] [Indexed: 12/21/2022]
Abstract
The discovery of chemotherapeutic agents for the treatment of cancer commonly uses cell proliferation assays in which cells grow as two-dimensional (2D) monolayers. Compounds identified using 2D monolayer assays often fail to advance during clinical development, most likely because these assays do not reproduce the cellular complexity of tumors and their microenvironment in vivo. The use of three-dimensional (3D) cellular systems have been explored as enabling more predictive in vitro tumor models for drug discovery. To date, small-scale screens have demonstrated that pharmacological responses tend to differ between 2D and 3D cancer cell growth models. However, the limited scope of screens using 3D models has not provided a clear delineation of the cellular pathways and processes that differentially regulate cell survival and death in the different in vitro tumor models. Here we sought to further understand the differences in pharmacological responses between cancer tumor cells grown in different conditions by profiling a large collection of 1912 chemotherapeutic agents. We compared pharmacological responses obtained from cells cultured in traditional 2D monolayer conditions with those responses obtained from cells forming spheres versus cells already in 3D spheres. The target annotation of the compound library screened enabled the identification of those key cellular pathways and processes that when modulated by drugs induced cell death in all growth conditions or selectively in the different cell growth models. In addition, we also show that many of the compounds targeting these key cellular functions can be combined to produce synergistic cytotoxic effects, which in many cases differ in the magnitude of their synergism depending on the cellular model and cell type. The results from this work provide a high-throughput screening framework to profile the responses of drugs both as single agents and in pairwise combinations in 3D sphere models of cancer cells.
Collapse
|
231
|
Coming-of-Age of Antibodies in Cancer Therapeutics. Trends Pharmacol Sci 2016; 37:1009-1028. [DOI: 10.1016/j.tips.2016.09.005] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 09/07/2016] [Accepted: 09/09/2016] [Indexed: 02/07/2023]
|
232
|
Chen L, Wilson K, Goldlust I, Mott BT, Eastman R, Davis MI, Zhang X, McKnight C, Klumpp-Thomas C, Shinn P, Simmons J, Gormally M, Michael S, Thomas CJ, Ferrer M, Guha R. mQC: A Heuristic Quality-Control Metric for High-Throughput Drug Combination Screening. Sci Rep 2016; 6:37741. [PMID: 27883049 PMCID: PMC5121902 DOI: 10.1038/srep37741] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 11/01/2016] [Indexed: 11/09/2022] Open
Abstract
Quality control (QC) metrics are critical in high throughput screening (HTS) platforms to ensure reliability and confidence in assay data and downstream analyses. Most reported HTS QC metrics are designed for plate level or single well level analysis. With the advent of high throughput combination screening there is a need for QC metrics that quantify the quality of combination response matrices. We introduce a predictive, interpretable, matrix-level QC metric, mQC, based on a mix of data-derived and heuristic features. mQC accurately reproduces the expert assessment of combination response quality and correctly identifies unreliable response matrices that can lead to erroneous or misleading characterization of synergy. When combined with the plate-level QC metric, Z', mQC provides a more appropriate determination of the quality of a drug combination screen. Retrospective analysis on a number of completed combination screens further shows that mQC is able to identify problematic screens whereas plate-level QC was not able to. In conclusion, our data indicates that mQC is a reliable QC filter that can be used to identify problematic drug combinations matrices and prevent further analysis on erroneously active combinations as well as for troubleshooting failed screens. The R source code of mQC is available at http://matrix.ncats.nih.gov/mQC.
Collapse
Affiliation(s)
- Lu Chen
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), Rockville, MD 20850, USA
| | - Kelli Wilson
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), Rockville, MD 20850, USA
| | - Ian Goldlust
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), Rockville, MD 20850, USA
| | - Bryan T. Mott
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), Rockville, MD 20850, USA
| | - Richard Eastman
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), Rockville, MD 20850, USA
| | - Mindy I. Davis
- National Institute of Allergy and Infectious Diseases (NIAID), Rockville, MD 20852, USA
| | - Xiaohu Zhang
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), Rockville, MD 20850, USA
| | - Crystal McKnight
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), Rockville, MD 20850, USA
| | - Carleen Klumpp-Thomas
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), Rockville, MD 20850, USA
| | - Paul Shinn
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), Rockville, MD 20850, USA
| | - John Simmons
- Laboratory of Cancer Biology and Genetics, National Cancer Institute (NCI), Bethesda, MD 20892, USA
| | - Mike Gormally
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), Rockville, MD 20850, USA
| | - Sam Michael
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), Rockville, MD 20850, USA
| | - Craig J. Thomas
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), Rockville, MD 20850, USA
| | - Marc Ferrer
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), Rockville, MD 20850, USA
| | - Rajarshi Guha
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), Rockville, MD 20850, USA
| |
Collapse
|
233
|
Wali VB, Langdon CG, Held MA, Platt JT, Patwardhan GA, Safonov A, Aktas B, Pusztai L, Stern DF, Hatzis C. Systematic Drug Screening Identifies Tractable Targeted Combination Therapies in Triple-Negative Breast Cancer. Cancer Res 2016; 77:566-578. [PMID: 27872098 DOI: 10.1158/0008-5472.can-16-1901] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 10/17/2016] [Accepted: 11/04/2016] [Indexed: 12/21/2022]
Abstract
Triple-negative breast cancer (TNBC) remains an aggressive disease without effective targeted therapies. In this study, we addressed this challenge by testing 128 FDA-approved or investigational drugs as either single agents or in 768 pairwise drug combinations in TNBC cell lines to identify synergistic combinations tractable to clinical translation. Medium-throughput results were scrutinized and extensively analyzed for sensitivity patterns, synergy, anticancer activity, and were validated in low-throughput experiments. Principal component analysis revealed that a fraction of all upregulated or downregulated genes of a particular targeted pathway could partly explain cell sensitivity toward agents targeting that pathway. Combination therapies deemed immediately tractable to translation included ABT-263/crizotinib, ABT-263/paclitaxel, paclitaxel/JQ1, ABT-263/XL-184, and paclitaxel/nutlin-3, all of which exhibited synergistic antiproliferative and apoptotic activity in multiple TNBC backgrounds. Mechanistic investigations of the ABT-263/crizotinib combination offering a potentially rapid path to clinic demonstrated RTK blockade, inhibition of mitogenic signaling, and proapoptotic signal induction in basal and mesenchymal stem-like TNBC. Our findings provide preclinical proof of concept for several combination treatments of TNBC, which offer near-term prospects for clinical translation. Cancer Res; 77(2); 566-78. ©2016 AACR.
Collapse
Affiliation(s)
- Vikram B Wali
- Department of Internal Medicine, Section of Medical Oncology, Yale School of Medicine, Yale University, New Haven, Connecticut. .,Yale Cancer Center, New Haven, Connecticut
| | - Casey G Langdon
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Matthew A Held
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - James T Platt
- Department of Internal Medicine, Section of Medical Oncology, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Gauri A Patwardhan
- Department of Internal Medicine, Section of Medical Oncology, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Anton Safonov
- Department of Internal Medicine, Section of Medical Oncology, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Bilge Aktas
- Department of Internal Medicine, Section of Medical Oncology, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Lajos Pusztai
- Department of Internal Medicine, Section of Medical Oncology, Yale School of Medicine, Yale University, New Haven, Connecticut.,Yale Cancer Center, New Haven, Connecticut
| | - David F Stern
- Yale Cancer Center, New Haven, Connecticut.,Department of Pathology, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Christos Hatzis
- Department of Internal Medicine, Section of Medical Oncology, Yale School of Medicine, Yale University, New Haven, Connecticut. .,Yale Cancer Center, New Haven, Connecticut
| |
Collapse
|
234
|
Ceribelli M, Hou ZE, Kelly PN, Huang DW, Wright G, Ganapathi K, Evbuomwan MO, Pittaluga S, Shaffer AL, Marcucci G, Forman SJ, Xiao W, Guha R, Zhang X, Ferrer M, Chaperot L, Plumas J, Jaffe ES, Thomas CJ, Reizis B, Staudt LM. A Druggable TCF4- and BRD4-Dependent Transcriptional Network Sustains Malignancy in Blastic Plasmacytoid Dendritic Cell Neoplasm. Cancer Cell 2016; 30:764-778. [PMID: 27846392 PMCID: PMC5175469 DOI: 10.1016/j.ccell.2016.10.002] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 07/27/2016] [Accepted: 10/03/2016] [Indexed: 12/21/2022]
Abstract
Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is an aggressive and largely incurable hematologic malignancy originating from plasmacytoid dendritic cells (pDCs). Using RNAi screening, we identified the E-box transcription factor TCF4 as a master regulator of the BPDCN oncogenic program. TCF4 served as a faithful diagnostic marker of BPDCN, and its downregulation caused the loss of the BPDCN-specific gene expression program and apoptosis. High-throughput drug screening revealed that bromodomain and extra-terminal domain inhibitors (BETis) induced BPDCN apoptosis, which was attributable to disruption of a BPDCN-specific transcriptional network controlled by TCF4-dependent super-enhancers. BETis retarded the growth of BPDCN xenografts, supporting their clinical evaluation in this recalcitrant malignancy.
Collapse
Affiliation(s)
- Michele Ceribelli
- Lymphoid Malignancies Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA; Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, MD 20892, USA
| | - Zhiying Esther Hou
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA
| | - Priscilla N Kelly
- Lymphoid Malignancies Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Da Wei Huang
- Lymphoid Malignancies Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - George Wright
- Biometric Research Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Karthik Ganapathi
- Laboratory of Pathology, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Moses O Evbuomwan
- Laboratory of Pathology, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Stefania Pittaluga
- Laboratory of Pathology, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Arthur L Shaffer
- Lymphoid Malignancies Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Guido Marcucci
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Stephen J Forman
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Wenming Xiao
- Division of Bioinformatics and Biostatistics, NCTR/FDA, Jefferson, AR 72079, USA
| | - Rajarshi Guha
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, MD 20892, USA
| | - Xiaohu Zhang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, MD 20892, USA
| | - Marc Ferrer
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, MD 20892, USA
| | - Laurence Chaperot
- R&D Laboratory, EFS Rhone-Alpes Grenoble, La Tronche 38701, France; Institute for Advanced Biosciences UGA, INSERM U1209, CNRS UMR 5309, Grenoble 38000, France
| | - Joel Plumas
- R&D Laboratory, EFS Rhone-Alpes Grenoble, La Tronche 38701, France; Institute for Advanced Biosciences UGA, INSERM U1209, CNRS UMR 5309, Grenoble 38000, France
| | - Elaine S Jaffe
- Laboratory of Pathology, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Craig J Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, MD 20892, USA
| | - Boris Reizis
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA; Department of Pathology, New York University School of Medicine, New York, NY 10016, USA.
| | - Louis M Staudt
- Lymphoid Malignancies Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
235
|
Approach to the diagnosis and treatment of high-grade B-cell lymphomas with MYC and BCL2 and/or BCL6 rearrangements. Blood 2016; 129:280-288. [PMID: 27821509 DOI: 10.1182/blood-2016-02-636316] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 10/25/2016] [Indexed: 12/19/2022] Open
Abstract
High-grade B-cell lymphomas (HGBLs) with MYC and BCL2 and/or BCL6 rearrangements, so-called "double-hit" lymphomas (HGBL-DH), are aggressive lymphomas that form a separate provisional entity in the 2016 revised World Health Organization Classification of Lymphoid Tumors. Fluorescence in situ hybridization (FISH) will be required to identify HGBL-DH and will reclassify a subset of diffuse large B-cell lymphomas (DLBCLs) and HGBLs with features intermediate between DLBCL and Burkitt lymphoma into this new category. Identifying patients with HGBL-DH is important because it may change clinical management. This poses a challenge for centers that may not be ready to handle the additional workload and financial burden associated with the increase in requests for FISH testing. Herein, we review the mechanisms of deregulation of these oncogenes. We identify the factors associated with a poor prognosis and those that can guide diagnostic testing. Restricting FISH analysis to the 10% of DLBCL patients who have a germinal center B-cell phenotype and coexpress MYC and BCL2 proteins would be cost-effective and would identify the subset of patients who are at highest risk of experiencing a relapse following conventional therapy. These patients may benefit from intensified chemotherapy regimens or, ideally, should enroll in clinical trials investigating novel regimens.
Collapse
|
236
|
Lee JHS, Vo TT, Fruman DA. Targeting mTOR for the treatment of B cell malignancies. Br J Clin Pharmacol 2016; 82:1213-1228. [PMID: 26805380 PMCID: PMC5061788 DOI: 10.1111/bcp.12888] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 01/20/2016] [Accepted: 01/21/2016] [Indexed: 12/12/2022] Open
Abstract
Mechanistic target of rapamycin (mTOR) is a serine/threonine kinase that functions as a key regulator of cell growth, division and survival. Many haematologic malignancies exhibit elevated or aberrant mTOR activation, supporting the launch of numerous clinical trials aimed at evaluating the potential of single agent mTOR-targeted therapies. While promising early clinical data using allosteric mTOR inhibitors (rapamycin and its derivatives, rapalogs) have suggested activity in a subset of haematologic malignancies, these agents have shown limited efficacy in most contexts. Whether the efficacy of these partial mTOR inhibitors might be enhanced by more complete target inhibition is being actively addressed with second generation ATP-competitive mTOR kinase inhibitors (TOR-KIs), which have only recently entered clinical trials. However, emerging preclinical data suggest that despite their biochemical advantage over rapalogs, TOR-KIs may retain a primarily cytostatic response. Rather, combinations of mTOR inhibition with other targeted therapies have demonstrated promising efficacy in several preclinical models. This review investigates the current status of rapalogs and TOR-KIs in B cell malignancies, with an emphasis on emerging preclinical evidence of synergistic combinations involving mTOR inhibition.
Collapse
Affiliation(s)
- Jong-Hoon Scott Lee
- Department of Molecular Biology & Biochemistry, University of California, Irvine, USA
| | - Thanh-Trang Vo
- Department of Molecular Biology & Biochemistry, University of California, Irvine, USA
| | - David A Fruman
- Department of Molecular Biology & Biochemistry, University of California, Irvine, USA.
| |
Collapse
|
237
|
Abstract
Recent advances in genomic sequencing and omics-based capabilities are uncovering tremendous therapeutic opportunities and rapidly transforming the field of cancer medicine. Molecularly targeted agents aim to exploit key tumor-specific vulnerabilities such as oncogenic or non-oncogenic addiction and synthetic lethality. Additionally, immunotherapies targeting the host immune system are proving to be another promising and complementary approach. Owing to substantial tumor genomic and immunologic complexities, combination strategies are likely to be required to adequately disrupt intricate molecular interactions and provide meaningful long-term benefit to patients. To optimize the therapeutic success and application of combination therapies, systematic scientific discovery will need to be coupled with novel and efficient clinical trial approaches. Indeed, a paradigm shift is required to drive precision medicine forward, from the traditional "drug-centric" model of clinical development in pursuit of small incremental benefits in large heterogeneous groups of patients, to a "strategy-centric" model to provide customized transformative treatments in molecularly stratified subsets of patients or even in individual patients. Crucially, to combat the numerous challenges facing combination drug development-including our growing but incomplete understanding of tumor biology, technical and informatics limitations, and escalating financial costs-aligned goals and multidisciplinary collaboration are imperative to collectively harness knowledge and fuel continual innovation.
Collapse
Affiliation(s)
- Daphne Day
- Drug Development Program, Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, M5G 2M9, Canada.,Department of Medicine, University of Toronto, Toronto, Ontario, M5S 1A8, Canada.,OICR Research Fellow, Ontario Institute for Cancer Research, Toronto, Ontario, M5G 0A3, Canada
| | - Lillian L Siu
- Drug Development Program, Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, M5G 2M9, Canada. .,Department of Medicine, University of Toronto, Toronto, Ontario, M5S 1A8, Canada.
| |
Collapse
|
238
|
Lee JS, Tang SS, Ortiz V, Vo TT, Fruman DA. MCL-1-independent mechanisms of synergy between dual PI3K/mTOR and BCL-2 inhibition in diffuse large B cell lymphoma. Oncotarget 2016; 6:35202-17. [PMID: 26460954 PMCID: PMC4742099 DOI: 10.18632/oncotarget.6051] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 09/24/2015] [Indexed: 11/25/2022] Open
Abstract
The PI3K/AKT/mTOR axis promotes survival and is a frequently mutated pathway in cancer. Yet, inhibitors targeting this pathway are insufficient to induce cancer cell death as single agents in some contexts, including diffuse large B cell lymphoma (DLBCL). In these situations, combinations with inhibitors targeting BCL-2 survival proteins (ABT-199 and ABT-263) may hold potential. Indeed, studies have demonstrated marked synergy in contexts where PI3K/mTOR inhibitors suppress expression of the pro-survival protein, MCL-1. In this study, we use BH3 profiling to confirm that BCL-2 and BCL-XL support survival following PI3K pathway inhibition, and that the dual PI3K/mTOR inhibitor BEZ235 strongly synergizes with BCL-2 antagonists in DLBCL. However, we identify an alternative mechanism of synergy between PI3K/mTOR and BCL-2 inhibitors, independent of MCL-1 down-regulation. Instead, we show that suppression of AKT activation by BEZ235 can induce the mitochondrial accumulation of pro-apoptotic BAD and BIM, and that expression of a constitutively active form of AKT prevents sensitization to BCL-2 antagonism. Thus, our work identifies an additional mechanism of synergy between PI3K pathway inhibitors and BCL-2 antagonists that strengthens the rationale for testing this combination in DLBCL.
Collapse
Affiliation(s)
- J Scott Lee
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | - Sarah S Tang
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | - Veronica Ortiz
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | - Thanh-Trang Vo
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | - David A Fruman
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| |
Collapse
|
239
|
Bogen D, Wei JS, Azorsa DO, Ormanoglu P, Buehler E, Guha R, Keller JM, Mathews Griner LA, Ferrer M, Song YK, Liao H, Mendoza A, Gryder BE, Sindri S, He J, Wen X, Zhang S, Shern JF, Yohe ME, Taschner-Mandl S, Shohet JM, Thomas CJ, Martin SE, Ambros PF, Khan J. Aurora B kinase is a potent and selective target in MYCN-driven neuroblastoma. Oncotarget 2016; 6:35247-62. [PMID: 26497213 PMCID: PMC4742102 DOI: 10.18632/oncotarget.6208] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 09/30/2015] [Indexed: 01/11/2023] Open
Abstract
Despite advances in multimodal treatment, neuroblastoma (NB) is often fatal for children with high-risk disease and many survivors need to cope with long-term side effects from high-dose chemotherapy and radiation. To identify new therapeutic targets, we performed an siRNA screen of the druggable genome combined with a small molecule screen of 465 compounds targeting 39 different mechanisms of actions in four NB cell lines. We identified 58 genes as targets, including AURKB, in at least one cell line. In the drug screen, aurora kinase inhibitors (nine molecules) and in particular the AURKB-selective compound, barasertib, were the most discriminatory with regard to sensitivity for MYCN-amplified cell lines. In an expanded panel of ten NB cell lines, those with MYCN-amplification and wild-type TP53 were the most sensitive to low nanomolar concentrations of barasertib. Inhibition of the AURKB kinase activity resulted in decreased phosphorylation of the known target, histone H3, and upregulation of TP53 in MYCN-amplified, TP53 wild-type cells. However, both wild-type and TP53 mutant MYCN-amplified cell lines arrested in G2/M phase upon AURKB inhibition. Additionally, barasertib induced endoreduplication and apoptosis. Treatment of MYCN-amplified/TP53 wild-type neuroblastoma xenografts resulted in profound growth inhibition and tumor regression. Therefore, aurora B kinase inhibition is highly effective in aggressive neuroblastoma and warrants further investigation in clinical trials.
Collapse
Affiliation(s)
- Dominik Bogen
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.,Children's Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| | - Jun S Wei
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - David O Azorsa
- Clinical Translational Research Division, Translational Genomics Research Institute (TGen), Scottsdale, AZ, USA
| | - Pinar Ormanoglu
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Eugen Buehler
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Rajarshi Guha
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Jonathan M Keller
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Lesley A Mathews Griner
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Marc Ferrer
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Young K Song
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Hongling Liao
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Arnulfo Mendoza
- Tumor and Metastasis Biology Section, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Berkley E Gryder
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sivasish Sindri
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jianbin He
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Xinyu Wen
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Shile Zhang
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - John F Shern
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Marielle E Yohe
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sabine Taschner-Mandl
- Children's Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| | - Jason M Shohet
- Texas Children's Cancer Center and Center for Cell and Gene Therapy, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Craig J Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Scott E Martin
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Peter F Ambros
- Children's Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| | - Javed Khan
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
240
|
Nagel D, Bognar M, Eitelhuber AC, Kutzner K, Vincendeau M, Krappmann D. Combinatorial BTK and MALT1 inhibition augments killing of CD79 mutant diffuse large B cell lymphoma. Oncotarget 2016; 6:42232-42. [PMID: 26540570 PMCID: PMC4747221 DOI: 10.18632/oncotarget.6273] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 09/30/2015] [Indexed: 11/25/2022] Open
Abstract
Survival of activated B cell-subtype (ABC) of diffuse large B cell lymphoma (DLBCL) is driven by chronic B cell receptor (BCR) signaling that activates the canonical NF-κB pathway. Inhibition of BTK by Ibrutinib has been shown to kill ABC DLBCL cells that carry activating mutations in the BCR adaptor CD79. However, mutations in BTK or in downstream components such as CARMA1/CARD11 can render lymphomas Ibrutinib resistant. Therefore, we assessed here the simultaneous inhibition of BTK and the protease MALT1 that acts downstream of CARMA1 and is essential for ABC DLBCL tumor growth. We show that in CD79 mutant cells BTK is a crucial upstream regulator of MALT1, but dispensable in CARMA1 mutant ABC DLBCL. Combined inhibition of BTK by Ibrutinib and MALT1 by S-Mepazine additively impaired MALT1 cleavage activity and expression of NF-κB pro-survival factors. Thereby, combinatorial Ibrutinib and S-Mepazine treatment enhanced killing of CD79 mutant ABC DLBCL cells. Moreover, while expression of oncogenic CARMA1 in CD79 mutant cells conferred Ibrutinib resistance, double mutant cells were still sensitive to MALT1 inhibition by S-Mepazine. Thus, based on the genetic background combinatorial BTK and MALT1 inhibition may improve effectiveness of therapeutic treatment and reduce the chances for the development of drug resistances.
Collapse
Affiliation(s)
- Daniel Nagel
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Miriam Bognar
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Andrea C Eitelhuber
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Kerstin Kutzner
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Michelle Vincendeau
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Daniel Krappmann
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
241
|
Di Veroli GY, Fornari C, Wang D, Mollard S, Bramhall JL, Richards FM, Jodrell DI. Combenefit: an interactive platform for the analysis and visualization of drug combinations. Bioinformatics 2016; 32:2866-8. [PMID: 27153664 PMCID: PMC5018366 DOI: 10.1093/bioinformatics/btw230] [Citation(s) in RCA: 517] [Impact Index Per Article: 57.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 04/05/2016] [Accepted: 04/20/2016] [Indexed: 01/10/2023] Open
Abstract
MOTIVATION Many drug combinations are routinely assessed to identify synergistic interactions in the attempt to develop novel treatment strategies. Appropriate software is required to analyze the results of these studies. RESULTS We present Combenefit, new free software tool that enables the visualization, analysis and quantification of drug combination effects in terms of synergy and/or antagonism. Data from combinations assays can be processed using classical Synergy models (Loewe, Bliss, HSA), as single experiments or in batch for High Throughput Screens. This user-friendly tool provides laboratory scientists with an easy and systematic way to analyze their data. The companion package provides bioinformaticians with critical implementations of routines enabling the processing of combination data. AVAILABILITY AND IMPLEMENTATION Combenefit is provided as a Matlab package but also as standalone software for Windows (http://sourceforge.net/projects/combenefit/). CONTACT Giovanni.DiVeroli@cruk.cam.ac.uk SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Giovanni Y Di Veroli
- CRUK Cambridge Institute, University of Cambridge, Cambridge, UK Early Clinical Development, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Cambridge, UK
| | - Chiara Fornari
- CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Dennis Wang
- Bioinformatics, Oncology Innovative Medicines, AstraZeneca, Cambridge, UK
| | - Séverine Mollard
- CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Jo L Bramhall
- CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
| | | | - Duncan I Jodrell
- CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|
242
|
Martinez NJ, Rai G, Yasgar A, Lea WA, Sun H, Wang Y, Luci DK, Yang SM, Nishihara K, Takeda S, Sagor M, Earnshaw I, Okada T, Mori K, Wilson K, Riggins GJ, Xia M, Grimaldi M, Jadhav A, Maloney DJ, Simeonov A. A High-Throughput Screen Identifies 2,9-Diazaspiro[5.5]Undecanes as Inducers of the Endoplasmic Reticulum Stress Response with Cytotoxic Activity in 3D Glioma Cell Models. PLoS One 2016; 11:e0161486. [PMID: 27570969 PMCID: PMC5003374 DOI: 10.1371/journal.pone.0161486] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 08/06/2016] [Indexed: 12/18/2022] Open
Abstract
The endoplasmic reticulum (ER) is involved in Ca2+ signaling and protein folding. ER Ca2+ depletion and accumulation of unfolded proteins activate the molecular chaperone GRP78 (glucose-regulated protein 78) which in turn triggers the ER stress response (ERSR) pathway aimed to restore ER homeostasis. Failure to adapt to stress, however, results in apoptosis. We and others have shown that malignant cells are more susceptible to ERSR-induced apoptosis than their normal counterparts, implicating the ERSR as a potential target for cancer therapeutics. Predicated on these findings, we developed an assay that uses a GRP78 biosensor to identify small molecule activators of ERSR in glioma cells. We performed a quantitative high-throughput screen (qHTS) against a collection of ~425,000 compounds and a comprehensive panel of orthogonal secondary assays was formulated for stringent compound validation. We identified novel activators of ERSR, including a compound with a 2,9-diazaspiro[5.5]undecane core, which depletes intracellular Ca2+ stores and induces apoptosis-mediated cell death in several cancer cell lines, including patient-derived and 3D cultures of glioma cells. This study demonstrates that our screening platform enables the identification and profiling of ERSR inducers with cytotoxic activity and advocates for characterization of these compound in in vivo models.
Collapse
Affiliation(s)
- Natalia J. Martinez
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, United States of America
| | - Ganesha Rai
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, United States of America
| | - Adam Yasgar
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, United States of America
| | - Wendy A. Lea
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, United States of America
| | - Hongmao Sun
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, United States of America
| | - Yuhong Wang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, United States of America
| | - Diane K. Luci
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, United States of America
| | - Shyh-Ming Yang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, United States of America
| | - Kana Nishihara
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, United States of America
- Department of Radiation Genetics, Graduate School of Medicine, Kyoto University, Yoshidakonoe, Sakyo, Kyoto 606–8501, Japan
| | - Shunichi Takeda
- Department of Neurosurgery, John Hopkins University, Baltimore, MD 21231, United States of America
| | - Mohiuddin Sagor
- Department of Radiation Genetics, Graduate School of Medicine, Kyoto University, Yoshidakonoe, Sakyo, Kyoto 606–8501, Japan
| | - Irina Earnshaw
- Department of Radiation Genetics, Graduate School of Medicine, Kyoto University, Yoshidakonoe, Sakyo, Kyoto 606–8501, Japan
| | - Tetsuya Okada
- Department of Biophysics, Graduate School of Science, Kyoto University, Kitashirakawa-Oiwake, Sakyo, Kyoto 606–8502, Japan
| | - Kazutoshi Mori
- Department of Biophysics, Graduate School of Science, Kyoto University, Kitashirakawa-Oiwake, Sakyo, Kyoto 606–8502, Japan
| | - Kelli Wilson
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, United States of America
- Department of Neurosurgery, John Hopkins University, Baltimore, MD 21231, United States of America
| | - Gregory J. Riggins
- Department of Neurosurgery, John Hopkins University, Baltimore, MD 21231, United States of America
| | - Menghang Xia
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, United States of America
| | - Maurizio Grimaldi
- Laboratory of Neuropharmacology, Department of Biochemistry and Molecular Biology, Southern Research Institute, Birmingham, AL 35205, United States of America
| | - Ajit Jadhav
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, United States of America
| | - David J. Maloney
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, United States of America
- * E-mail: (AS); (DJM)
| | - Anton Simeonov
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, United States of America
- * E-mail: (AS); (DJM)
| |
Collapse
|
243
|
SYGNALing a Red Light for Glioblastoma. Cell Syst 2016; 3:118-120. [PMID: 27559923 DOI: 10.1016/j.cels.2016.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
A new multiomic network inference pipeline, SYGNAL, integrates patient data with mechanistically accurate transcriptional regulatory networks to predict drug combinations with synergistic anti-proliferative effects on glioblastoma multiforme.
Collapse
|
244
|
Antignani A, Mathews Griner L, Guha R, Simon N, Pasetto M, Keller J, Huang M, Angelus E, Pastan I, Ferrer M, FitzGerald DJ, Thomas CJ. Chemical Screens Identify Drugs that Enhance or Mitigate Cellular Responses to Antibody-Toxin Fusion Proteins. PLoS One 2016; 11:e0161415. [PMID: 27556570 PMCID: PMC4996465 DOI: 10.1371/journal.pone.0161415] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 08/04/2016] [Indexed: 01/03/2023] Open
Abstract
The intersection of small molecular weight drugs and antibody-based therapeutics is rarely studied in large scale. Both types of agents are currently part of the cancer armamentarium. However, very little is known about how to combine them in optimal ways. Immunotoxins are antibody-toxin gene fusion proteins engineered to target cancer cells via antibody binding to surface antigens. For fusion proteins derived from Pseudomonas exotoxin (PE), potency relies on the enzymatic domain of the toxin which catalyzes the ADP-ribosylation of EF2 causing inhibition of protein synthesis leading to cell death. Candidate immunotoxins have demonstrated clear value in clinical trials but generally have not been curative as single agents. Therefore we undertook three screens to discover effective combinations that could act synergistically. From the MIPE-3 library of compounds we identified various enhancers of immunotoxin action and at least one major class of inhibitor. Follow-up experiments confirmed the screening data and suggested that immunotoxins when administered with everolimus or nilotinib exhibit favorable combinatory activity and would be candidates for preclinical development. Mechanistic studies revealed that everolimus-immunotoxin combinations acted synergistically on elements of the protein synthetic machinery, including S61 kinase and 4E-BP1 of the mTORC1 pathway. Conversely, PARP inhibitors antagonized immunotoxins and also blocked the toxicity due to native ADP-ribosylating toxins. Thus, our goal of investigating a chemical library was justified based on the identification of several approved compounds that could be developed preclinically as ‘enhancers’ and at least one class of mitigator to be avoided.
Collapse
Affiliation(s)
- Antonella Antignani
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892–4264, United States of America
| | - Lesley Mathews Griner
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, 20850, United States of America
| | - Rajarshi Guha
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, 20850, United States of America
| | - Nathan Simon
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892–4264, United States of America
| | - Matteo Pasetto
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892–4264, United States of America
| | - Jonathan Keller
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, 20850, United States of America
| | - Manjie Huang
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892–4264, United States of America
| | - Evan Angelus
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892–4264, United States of America
| | - Ira Pastan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892–4264, United States of America
| | - Marc Ferrer
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, 20850, United States of America
| | - David J. FitzGerald
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892–4264, United States of America
- * E-mail: (DF); (CJT)
| | - Craig J. Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, 20850, United States of America
- * E-mail: (DF); (CJT)
| |
Collapse
|
245
|
Yasuhiro T, Sawada W, Klein C, Kozaki R, Hotta S, Yoshizawa T. Anti-tumor efficacy study of the Bruton's tyrosine kinase (BTK) inhibitor, ONO/GS-4059, in combination with the glycoengineered type II anti-CD20 monoclonal antibody obinutuzumab (GA101) demonstrates superior in vivo efficacy compared to ONO/GS-4059 in combination with rituximab. Leuk Lymphoma 2016; 58:699-707. [PMID: 27684575 DOI: 10.1080/10428194.2016.1201567] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The activated B-cell diffuse large B-cell-like lymphoma (ABC-DLBCL) correlates with poor prognosis. The B-cell receptor signaling pathway is known to be dysregulated in NHL/CLL and given BTK is a downstream mediator of BCR signaling, BTK constitutes an interesting and obvious therapeutic target. Given the high potency and selectivity of the BTK inhibitor, ONO/GS-4059, it was hypothesized that, the anti-tumor activity of ONO/GS-4059 could be further enhanced by combining it with the anti-CD20 Abs, rituximab (RTX) or obinutuzumab (GA101). ONO/GS-4059 combined with GA101 or RTX was significantly better than the respective monotherapy with tumor growth inhibition (TGI) of 90% for the GA101 combination and 86% for the RTX combination. In contrast, ibrutinib (PCI-32765) combined with RTX did not result in improved efficacy compared with respective monotherapy. Taken together these data indicate that the combination of ONO/GS-4059 with rituximab and particularly obinutuzumab may be an effective treatment for ABC-DLBCL.
Collapse
Affiliation(s)
| | - Wako Sawada
- a Ono Pharmaceutical Co, Ltd , Osaka , Japan
| | - Christian Klein
- b Roche Pharmaceutical Research & Early Development, Roche Innovation Center Zurich , Schlieren , Switzerland
| | | | | | | |
Collapse
|
246
|
Zhang B, Wang T, Yang S, Xiao Y, Song Y, Zhang N, Garg S. Development and evaluation of oxaliplatin and irinotecan co-loaded liposomes for enhanced colorectal cancer therapy. J Control Release 2016; 238:10-21. [PMID: 27432750 DOI: 10.1016/j.jconrel.2016.07.022] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 06/29/2016] [Accepted: 07/13/2016] [Indexed: 01/22/2023]
Abstract
Drug combinations are widely employed in chemotherapy for colorectal cancer treatment. However, traditional cocktail combination in clinic causes the uncertainty of the treatment, owing to varying pharmacokinetics of different drugs. The aim of this study was to design co-loaded liposomes to achieve the synchronised delivery and release. Oxaliplatin and irinotecan hydrochloride, as one of recommended combination schemes for the treatment of colorectal cancer in clinic, were co-loaded into the liposomes. The particle sizes of the liposomes were <200nm with uniform size distribution. In vitro release study showed that both drugs could be synchronously released from the liposomes, which means the optimized synergistic ratio of two drugs could be achieved. In vitro cellular uptake revealed that co-loaded liposomes could efficiently deliver different drugs into the same cells, indicating their potential as carriers for enhancing the cancer therapy. CLSM images of cryo-sections for in vivo co-delivery study also revealed that co-loaded liposomes had superior ability to co-deliver both the cargoes into the same tumor cells. Besides, in vivo NIRF imaging indicated that the liposomes could increase the drug accumulation in tumor compared with free drug. In vitro cytotoxicity evaluation demonstrated that co-loaded liposomes exhibited higher cytotoxicity than the mixture of single loaded liposomes in both CT-26 and HCT-116 cells. Furthermore, co-loaded liposomes also presented superior anti-tumor activity in CT-26 bearing BALB/c mice. In vivo safety assessment demonstrated that liposomes had lower toxicities than their solution formulations. These results indicated that oxaliplatin and irinotecan hydrochloride co-loaded liposomes would be an efficient formulation for improving colorectal cancer therapy with potential clinical applications.
Collapse
Affiliation(s)
- Bo Zhang
- School of Pharmaceutical Science, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong Province 250012, China
| | - Tianqi Wang
- School of Pharmaceutical Science, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong Province 250012, China
| | - Shaomei Yang
- School of Pharmaceutical Science, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong Province 250012, China
| | - Yanan Xiao
- School of Pharmaceutical Science, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong Province 250012, China
| | - Yunmei Song
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Na Zhang
- School of Pharmaceutical Science, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong Province 250012, China.
| | - Sanjay Garg
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia 5001, Australia
| |
Collapse
|
247
|
|
248
|
Manzoni D, Catallo R, Chebel A, Baseggio L, Michallet AS, Roualdes O, Magaud JP, Salles G, Ffrench M. The ibrutinib B-cell proliferation inhibition is potentiated in vitro by dexamethasone: Application to chronic lymphocytic leukemia. Leuk Res 2016; 47:1-7. [PMID: 27235717 DOI: 10.1016/j.leukres.2016.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 05/05/2016] [Accepted: 05/09/2016] [Indexed: 01/26/2023]
Abstract
New B-cell receptor-targeted therapies such as ibrutinib, a Bruton tyrosine kinase inhibitor, are now proposed for lymphoid pathologies. The putative benefits of its combination with glucocorticoids were evaluated here. We compared the effects of dexamethasone (DXM), ibrutinib and their in vitro combination on proliferation and metabolic stress markers in stimulated normal B-lymphocytes and in malignant lymphocytes from chronic lymphocytic leukemia (CLL) patients. In both cellular models, cell cycle progression was globally inhibited by DXM and/or ibrutinib. This inhibition was significantly amplified by DXM addition to ibrutinib and was related to a significant decrease in the expression of the cell cycle regulatory proteins CDK4 and cyclin E. Apoptosis increased especially with DXM/ibrutinib combination and was associated with a significant decrease in Mcl-1 expression. Treatment effects on metabolic stress were evaluated by DNA damage recognition after 53BP1 foci labeling. The percentage of cells with more than five 53BP1 foci decreased significantly with ibrutinib in normal and CLL lymphocytes. This decrease was strongly reinforced, in CLL, by DXM addition. Our data indicated that, in vitro, DXM potentiated antiproliferative effects of ibrutinib and decreased DNA damage in lymphoid B-cells. Thus their combination may be proposed for CLL treatment.
Collapse
Affiliation(s)
- Delphine Manzoni
- Laboratory "Proliférations B indolentes", Université Claude Bernard Lyon 1, CNRS UMR 5239-Ecole Normale Supérieure (ENS)-Hospices Civil de Lyon (HCL), Faculté de Médecine Lyon Sud, France; Hematology laboratory, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), France
| | - Régine Catallo
- Laboratory "Proliférations B indolentes", Université Claude Bernard Lyon 1, CNRS UMR 5239-Ecole Normale Supérieure (ENS)-Hospices Civil de Lyon (HCL), Faculté de Médecine Lyon Sud, France
| | - Amel Chebel
- Laboratory "Proliférations B indolentes", Université Claude Bernard Lyon 1, CNRS UMR 5239-Ecole Normale Supérieure (ENS)-Hospices Civil de Lyon (HCL), Faculté de Médecine Lyon Sud, France
| | - Lucile Baseggio
- Laboratory "Proliférations B indolentes", Université Claude Bernard Lyon 1, CNRS UMR 5239-Ecole Normale Supérieure (ENS)-Hospices Civil de Lyon (HCL), Faculté de Médecine Lyon Sud, France; Hematology laboratory, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), France
| | - Anne-Sophie Michallet
- Hematology department, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), France
| | - Olivier Roualdes
- Laboratory "Proliférations B indolentes", Université Claude Bernard Lyon 1, CNRS UMR 5239-Ecole Normale Supérieure (ENS)-Hospices Civil de Lyon (HCL), Faculté de Médecine Lyon Sud, France; Hematology laboratory, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), France
| | - Jean-Pierre Magaud
- Hematology laboratory, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), France
| | - Gilles Salles
- Laboratory "Proliférations B indolentes", Université Claude Bernard Lyon 1, CNRS UMR 5239-Ecole Normale Supérieure (ENS)-Hospices Civil de Lyon (HCL), Faculté de Médecine Lyon Sud, France; Hematology department, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), France
| | - Martine Ffrench
- Laboratory "Proliférations B indolentes", Université Claude Bernard Lyon 1, CNRS UMR 5239-Ecole Normale Supérieure (ENS)-Hospices Civil de Lyon (HCL), Faculté de Médecine Lyon Sud, France; Hematology laboratory, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), France; Hematology department, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), France.
| |
Collapse
|
249
|
Li Y, Bouchlaka MN, Wolff J, Grindle KM, Lu L, Qian S, Zhong X, Pflum N, Jobin P, Kahl BS, Eickhoff JC, Wuerzberger-Davis SM, Miyamoto S, Thomas CJ, Yang DT, Capitini CM, Rui L. FBXO10 deficiency and BTK activation upregulate BCL2 expression in mantle cell lymphoma. Oncogene 2016; 35:6223-6234. [PMID: 27157620 DOI: 10.1038/onc.2016.155] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 02/29/2016] [Accepted: 03/11/2016] [Indexed: 12/17/2022]
Abstract
Targeting Bruton tyrosine kinase (BTK) by ibrutinib is an effective treatment for patients with relapsed/refractory mantle cell lymphoma (MCL). However, both primary and acquired resistance to ibrutinib have developed in a significant number of these patients. A combinatory strategy targeting multiple oncogenic pathways is critical to enhance the efficacy of ibrutinib. Here, we focus on the BCL2 anti-apoptotic pathway. In a tissue microarray of 62 MCL samples, BCL2 expression positively correlated with BTK expression. Increased levels of BCL2 were shown to be due to a defect in protein degradation because of no or little expression of the E3 ubiquitin ligase FBXO10, as well as transcriptional upregulation through BTK-mediated canonical nuclear factor-κB activation. RNA-seq analysis confirmed that a set of anti-apoptotic genes (for example, BCL2, BCL-XL and DAD1) was downregulated by BTK short hairpin RNA. The downregulated genes also included those that are critical for B-cell growth and proliferation, such as BCL6, MYC, PIK3CA and BAFF-R. Targeting BCL2 by the specific inhibitor ABT-199 synergized with ibrutinib in inhibiting growth of both ibrutinib-sensitive and -resistant cancer cells in vitro and in vivo. These results suggest co-targeting of BTK and BCL2 as a new therapeutic strategy in MCL, especially for patients with primary resistance to ibrutinib.
Collapse
Affiliation(s)
- Y Li
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - M N Bouchlaka
- Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - J Wolff
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - K M Grindle
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - L Lu
- Wisconsin Institute for Discovery and Laboratory of Genetics, University of Wisconsin, Madison, WI, USA
| | - S Qian
- Wisconsin Institute for Discovery and Laboratory of Genetics, University of Wisconsin, Madison, WI, USA
| | - X Zhong
- Wisconsin Institute for Discovery and Laboratory of Genetics, University of Wisconsin, Madison, WI, USA
| | - N Pflum
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - P Jobin
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - B S Kahl
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - J C Eickhoff
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - S M Wuerzberger-Davis
- Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - S Miyamoto
- Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - C J Thomas
- Division of Preclinical Innovation, National Institutes of Health Chemical Genomics Center, National Center for Advancing Translational Sciences, Bethesda, MD, USA
| | - D T Yang
- Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - C M Capitini
- Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - L Rui
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
250
|
Guha R, Mathews Griner LA, Keller JM, Zhang X, Fitzgerald D, Antignani A, Pastan I, Thomas CJ, Ferrer M. Ranking Differential Drug Activities from Dose-Response Synthetic Lethality Screens. ACTA ACUST UNITED AC 2016; 21:942-55. [PMID: 27112173 DOI: 10.1177/1087057116644890] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 03/28/2016] [Indexed: 12/16/2022]
Abstract
Synthetic lethal screens are used to discover new combination treatments for cancer. In traditional high-throughput synthetic lethal screens, compounds are tested at a single dose, and hit selection is based on threshold activity values from the variance of the efficacy of the compounds tested. The limitation of the single-dose screening for synthetic lethal screens is that it does not allow for the robust detection of differential activities from compound collections with a broad range of potencies and efficacies. There is therefore a need to develop screening approaches that enable the identification of compounds with synthetic lethal effects based on changes in both potency and efficacy. Here we describe the implementation of a dose response-based synthetic lethal screen to find drugs that enhance or mitigate the cytotoxic effect of an immunotoxin protein (HA22). We developed a data analysis framework for the selection of compounds with enhancing or mitigating cytotoxic activities based on the use of dose-response parameters. The data analysis framework includes an ensemble ranking approach that allows the use of multiple dose-response parameters in a nonparametric fashion. Quantitative high-throughput screening (HTS) enables the identification of compounds with synthetic lethal activity not identified by single-dose HTS.
Collapse
Affiliation(s)
- Rajarshi Guha
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Lesley A Mathews Griner
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Jonathan M Keller
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Xiaohu Zhang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - David Fitzgerald
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Antonella Antignani
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ira Pastan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Craig J Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Marc Ferrer
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|