28 results on '"Claudia M. Espitia"'
Search Results
2. Resistance to histone deacetylase inhibitors confers hypersensitivity to oncolytic reovirus therapy
- Author
-
Shariful Islam, Jennifer S. Carew, Claudia M. Espitia, Steffan T. Nawrocki, and Daniel O. Persky
- Subjects
Oncolytic Virotherapy ,Vorinostat ,Lymphoid Neoplasia ,business.industry ,Hematology ,Drug resistance ,Lymphoma, T-Cell ,Oncolytic virus ,Romidepsin ,Histone Deacetylase Inhibitors ,Histones ,chemistry.chemical_compound ,chemistry ,Panobinostat ,Cell Line, Tumor ,Reolysin ,Cancer research ,medicine ,Humans ,Histone deacetylase ,business ,Belinostat ,medicine.drug - Abstract
Despite the promising antilymphoma activity of histone deacetylase (HDAC) inhibitors as a drug class, resistance is a significant clinical issue. Elucidating the molecular mechanisms driving HDAC inhibitor resistance and/or the specific targets that are altered in drug-resistant cells may facilitate the development of strategies that overcome drug resistance and are more effective for refractory patients. We generated novel T-cell lymphoma (TCL) cell line models of acquired resistance to the HDAC inhibitor belinostat to identify potential effective therapies. Belinostat-resistant cells displayed significant cross-resistance to other HDAC inhibitors including romidepsin, panobinostat, and vorinostat. Consistent with a lack of sensitivity to HDAC inhibitors, the resistant cells failed to induce increased acetylated histones. Drug-resistant cells featured significantly decreased expression of the key antiviral mediators IRF1 and STAT1. On the basis of these findings, we investigated the efficacy of the clinical formulation of reovirus (Reolysin) in parental and drug-resistant models. Our investigation revealed that HDAC inhibitor–resistant cells displayed enhanced vulnerability to reovirus replication and cell death in both in vitro and in vivo models compared with their parental counterparts. Importantly, Reolysin also significantly increased the antilymphoma activity of belinostat in HDAC inhibitor–resistant cells. Our data demonstrate that Reolysin alone or in combination with belinostat is a novel therapeutic strategy to treat TCL patients who develop resistance to HDAC inhibitors.
- Published
- 2020
3. Moving beyond hydroxychloroquine: the novel lysosomal autophagy inhibitor ROC-325 shows significant potential in preclinical studies
- Author
-
Wei Wang, Claudia M. Espitia, Jennifer S. Carew, Steffan T. Nawrocki, and Trace M. Jones
- Subjects
Cancer Research ,Myeloid ,Antineoplastic Agents ,lcsh:RC254-282 ,Mice ,Text mining ,Cell Line, Tumor ,Autophagy ,medicine ,Carcinoma ,Animals ,Carcinoma, Renal Cell ,business.industry ,Hydroxychloroquine ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Research Highlight ,Kidney Neoplasms ,Leukemia, Myeloid, Acute ,Leukemia ,medicine.anatomical_structure ,Oncology ,Azacitidine ,Cancer research ,Lysosomes ,business ,medicine.drug - Published
- 2019
- Full Text
- View/download PDF
4. The Clinical Oncolytic Reovirus Formulation Reolysin Synergistically Augments the Anti-Leukemic Activity of Azacitidine
- Author
-
Rayna L Schwartz, Daniel O. Persky, Jennifer S. Carew, Claudia M. Espitia, and Steffan T. Nawrocki
- Subjects
business.industry ,Immunology ,Azacitidine ,Reolysin ,Cancer research ,Medicine ,Cell Biology ,Hematology ,business ,Biochemistry ,Oncolytic virus ,medicine.drug - Abstract
Despite the recent development of new agents for acute myeloid leukemia (AML) therapy, novel approaches are still needed for patients that do not benefit sufficiently from existing regimens. Reolysin (Pelareorep) is a proprietary clinical formulation of the naturally occurring oncolytic reovirus that is non-pathogenic and preferentially replicates in cancer cells, but not in normal tissue. Although Reolysin has been investigated in over 30 adult clinical trials and is very well tolerated when given alone and in combination with chemotherapy, it has never been evaluated for AML therapy. Our major goal was to determine the efficacy and mechanism of action of Reolysin alone and in combination with azacitidine in AML models and primary patient specimens. Reolysin exhibited dose-dependent effects against AML cells with respect to the reduction in leukemia cell viability and induction of apoptosis in all 8 cell lines evaluated. The combination of Reolysin with azacitidine yielded synergistic benefit across all cell lines that was dramatically superior to single agent treatments (p Disclosures No relevant conflicts of interest to declare.
- Published
- 2021
- Full Text
- View/download PDF
5. Oncolytic reovirus inhibits angiogenesis through induction of CXCL10/IP-10 and abrogation of HIF activity in soft tissue sarcomas
- Author
-
Monica M. Mita, Claudia M. Espitia, Steffan T. Nawrocki, Jennifer S. Carew, Weiguo Zhao, and Alain C. Mita
- Subjects
0301 basic medicine ,Angiogenesis ,reovirus ,03 medical and health sciences ,chemistry.chemical_compound ,angiogenesis ,0302 clinical medicine ,Reolysin ,medicine ,CXCL10 ,Sunitinib ,business.industry ,HIF-1 ,Temsirolimus ,3. Good health ,Oncolytic virus ,Vascular endothelial growth factor ,030104 developmental biology ,Oncology ,chemistry ,Hypoxia-inducible factors ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,reolysin ,business ,medicine.drug ,Research Paper - Abstract
The tumor-selective viral replication capacity and pro-apoptotic effects of oncolytic reovirus have been reported to be dependent on the presence of an activated RAS pathway in several solid tumor types. However, the mechanisms of selective anticancer efficacy of the reovirus-based formulation for cancer therapy (Reolysin, pelareorep) have not been rigorously studied in soft tissue sarcomas (STS). Here we report that Reolysin triggered a striking induction of the anti-angiogenic chemokine interferon-γ-inducible protein 10 (IP-10)/CXCL10 (CXC chemokine ligand 10) in both wild type and RAS mutant STS cells. Further analysis determined that Reolysin treatment possessed significant anti-angiogenic activity irrespective of RAS status. In addition to CXCL10 induction, Reolysin dramatically downregulated the expression of hypoxia inducible factor (HIF)-1α, HIF-2α and inhibited vascular endothelial growth factor (VEGF) secretion. CXCL10 antagonism significantly diminished the anti-angiogenic effects of Reolysin indicating that it is a key driver of this phenomenon. Xenograft studies demonstrated that Reolysin significantly improved the anticancer activity of the anti-angiogenic agents sunitinib, temsirolimus, and bevacizumab in a manner that was associated with increased CXCL10 levels. This effect was most pronounced following treatment with Reolysin in combination with temsirolimus. Further analysis in additional sarcoma xenograft models confirmed the significant increase in CXCL10 and increased anticancer activity of this combination. Our collective results demonstrate that Reolysin possesses CXCL10-driven anti-angiogenic activity in sarcoma models, which can be harnessed to enhance the anticancer activity of temsirolimus and other agents that target the tumor vasculature.
- Published
- 2017
6. Oncolytic reovirus sensitizes multiple myeloma cells to anti-PD-L1 therapy
- Author
-
Jennifer S. Carew, Valeria Visconte, Kaijin Wu, Christine M. Calton, Claudia M. Espitia, Weiguo Zhao, Kevin R. Kelly, Steffan T. Nawrocki, and Faiz Anwer
- Subjects
PD-L1 ,0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,Plasma Cells ,reovirus ,B7-H1 Antigen ,Article ,Targeted therapy ,Mice ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,Reolysin ,Animals ,Humans ,Medicine ,oncolytic virus ,B-Lymphocytes ,Clinical Trials as Topic ,biology ,business.industry ,Bortezomib ,Hematology ,Immune checkpoint ,3. Good health ,Oncolytic virus ,multiple myeloma ,Oncolytic Viruses ,030104 developmental biology ,Oncology ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Antibody ,business ,medicine.drug - Abstract
Adaptive resistance mediated by inhibitory ligands such as programmed death 1 ligand (PD-L1) has emerged as an important mechanism of malignant cell survival. This has spurred the development of new agents that disrupt the PD-L1/PD-1 immune checkpoint. Analysis of patient specimens from clinical trials of novel immune checkpoint inhibitors indicates that high basal expression of PD-L1 on tumor cells may predict sensitivity to and be necessary to elicit significant clinical benefit from this drug class. Notably, many multiple myeloma (MM) cell lines and primary CD138+ cells from MM patients do not overexpress PD-L1 compared to normal plasma cells and this may preclude patients with MM from optimally benefitting from immune checkpoint inhibitor therapy. These data suggest that strategies that transiently increase PD-L1 levels could potentially sensitize malignant cells with low PD-L1 expression to anti-PD-1/PD-L1 blockade. The oncolytic reovirus-based anticancer agent Reolysin is known to have significant immunomodulatory effects and has demonstrated promising preclinical efficacy in MM models and favorable safety and tolerability in early MM clinical trials. We demonstrated that Reolysin selectively replicates in MM cells and possesses significant activity in preclinical in vitro and in vivo MM models. These findings established the framework for an ongoing investigator-initiated phase 1b clinical study of Reolysin in combination with bortezomib and dexamethasone in patients with relapsed/refractory MM. Recent gene ontology analyses of RPMI-8226 and U266 MM cells treated with Reolysin revealed that reovirus exposure triggers a highly significant transient increase in CD274 (PD-L1) in MM cell lines. Reolysin-mediated PD-L1 upregulation was confirmed by immunoblotting, qRT-PCR, and flow cytometric analyses in MM cell lines and primary patient specimens treated with Reolysin. Increased PD-L1 expression was also detected by immunohistochemistry in MM tumor samples collected from mice treated with Reolysin. A comparison of the anti-MM effects achieved by live reovirus versus UV-inactivated reovirus demonstrated that live reovirus is required to decrease MM cell viability and upregulate PD-L1 expression. Our data demonstrate proof of concept that reovirus infection and replication in MM cells can efficiently and selectively upregulate PD-L1 levels in malignant cells with low target expression. We hypothesized that Reolysin treatment could be used as a precision priming strategy to potentiate the anti-MM efficacy of PD-1/PD-L1 targeted therapy by promoting myeloma immune recognition and PD-L1 upregulation. To investigate this therapeutic approach, 5TGM1-luc murine MM cells were injected IV into immunocompetent mice to generate MM bone disease. After disease was established, mice were randomized into groups and treated with vehicle, Reolysin (5 x 108 TCID50, Q7D), murine anti-PD-L1 antibody (200 mg/mouse, Q2D) or the combination for 5 weeks. Mice treated with the combination demonstrated decreased disease burden as measured by bioluminescent imaging and also showed reduced IgG2bk levels (specific IgG secreted by 5TGM1 cells) by ELISA. Importantly, the combination also led to increased overall animal survival compared to vehicle control and either single agent treatment (P Disclosures Kelly: Amgen: Honoraria; Abbvie: Honoraria; Pharmacyclics: Honoraria; Jannsen: Honoraria.
- Published
- 2017
- Full Text
- View/download PDF
7. Heightened JNK Activation and Reduced XIAP Levels Promote TRAIL and Sunitinib-Mediated Apoptosis in Colon Cancer Models
- Author
-
Jennifer S. Carew, Robbert H. Cool, Claudia M. Espitia, Steffan T. Nawrocki, Steven de Jong, Devalingam Mahalingam, Francis J. Giles, Targeted Gynaecologic Oncology (TARGON), and Guided Treatment in Optimal Selected Cancer Patients (GUTS)
- Subjects
0301 basic medicine ,Cancer Research ,sunitinib ,TRAIL ,Apoptosis ,Inhibitor of apoptosis ,urologic and male genital diseases ,lcsh:RC254-282 ,Article ,Small hairpin RNA ,03 medical and health sciences ,0302 clinical medicine ,XIAP ,medicine ,Sunitinib ,Kinase ,Chemistry ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,3. Good health ,030104 developmental biology ,Oncology ,colon cancer ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Tumor necrosis factor alpha ,medicine.drug - Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a potent inducer of apoptosis that may be a promising agent in cancer therapy due to its selectivity toward tumor cells. However, many cancer cells are resistant to TRAIL due to defects in apoptosis signaling or activation of survival pathways. We hypothesized that a disruption of pro-survival signaling cascades with the multi-tyrosine kinase inhibitor sunitinib and would be an effective strategy to enhance TRAIL-mediated apoptosis. Here we demonstrate that sunitinib significantly augments the anticancer activity of TRAIL in models of colon cancer. The therapeutic benefit of the TRAIL/sunitinib combination was associated with increased apoptosis marked by enhanced caspase-3 cleavage and DNA fragmentation. Overexpression of the anti-apoptotic factor B-cell lymphoma 2 (BCL-2) in HCT116 cells reduced TRAIL/sunitinib-mediated apoptosis, further supporting that sunitinib enhances the anticancer activity of TRAIL via augmented apoptosis. Analysis of pro-survival factors identified that the combination of TRAIL and sunitinib significantly downregulated the anti-apoptotic protein X-linked inhibitor of apoptosis protein (XIAP) through a c-Jun N-terminal kinase (JNK)-mediated mechanism. Short hairpin RNA (shRNA)-mediated knockdown of JNK confirmed its key role in the regulation of sensitivity to this combination as cells with suppressed JNK expression exhibited significantly reduced TRAIL/sunitinib-mediated apoptosis. Importantly, the therapeutic benefit of the TRAIL/sunitinib combination was validated in the HCT116-Luc and HCT15 colon cancer xenograft models, which both demonstrated significant anti-tumor activity in response to combination treatment. Collectively, our data demonstrate that sunitinib enhances TRAIL-mediated apoptosis by heightened JNK activation, diminished XIAP levels, and augmented apoptosis.
- Published
- 2019
8. Abstract 5211: Targeting Neddylation synergistically augments the activity of cisplatin in head and neck cancer models to yield long-term survival by modulating key DNA damage and redox pathways
- Author
-
Jennifer S. Carew, Steffan T. Nawrocki, Claudia M. Espitia, and Trace M. Jones
- Subjects
Cisplatin ,Cancer Research ,DNA damage ,business.industry ,Cancer ,Cell cycle ,medicine.disease ,Pevonedistat ,Oncology ,Apoptosis ,Cancer research ,medicine ,DNA fragmentation ,Neddylation ,business ,medicine.drug - Abstract
Head and neck cancer is a broad term for neoplasms of the epithelial lining of the anatomical sites of the upper aerodigestive tract. Very few novel therapeutics have been developed for the treatment of advanced head and neck cancer with cisplatin continuing to be a key standard of care agent. Innovative and more effective therapeutic strategies are urgently needed. The inappropriate loss of several NEDD8-cullin-RING ubiquitin ligase (CRL) targets has been linked to cancer progression and drug resistance. Our data show that overexpression of NEDD8 activating enzyme (NAE), a key proximal regulator of the NEDDylation cascade, is correlated with inferior survival in head and neck cancers suggesting that this pathway may be a promising new therapeutic target. We hypothesized that disrupting NEDDylation with the first-in-class NAE inhibitor pevonedistat (MLN4924, PEV) would be an effective new approach for the treatment of head and neck cancers. Initial testing demonstrated that PEV potently impaired the NEDDylation cascade as evidenced by decreased NEDDylated cullins and upregulation of the CRL target cell cycle inhibitors, p21 and p27. In addition, PEV induced a dramatic G2 cell cycle arrest, polyploidy, and activation of the DNA damage response. PEV treatment also potently decreased cell viability and induced apoptosis in a panel of head and neck cancer cell lines with diverse genetic features. Given the potent DNA damage-related aspects of PEV's mechanism of action, we hypothesized that it would be a rational combination partner for standard cisplatin therapy. Consistent with this hypothesis, co-treatment with PEV and cisplatin promoted synergistic levels of DNA damage as measured by comet assay, an accumulation of gamma-H2AX, and quantification of DNA fragmentation. The combination also cooperated to promote redox alterations that were necessary for apoptosis induction and linked to the DNA damaging effects of both drugs. Importantly, synergistic anticancer activity was observed in cell lines from multiple anatomical sites, suggesting that this combination has broad efficacy across many subtypes of head and neck cancer. To further evaluate the therapeutic benefit of the PEV and cisplatin combination, we determined its safety and efficacy in a head and neck cancer xenograft model. Consistent with our in vitro data, the combination significantly decreased tumor proliferation, induced apoptosis, and augmented DNA damage in vivo. While both PEV and cisplatin displayed significant activity as monotherapies, combination treatment resulted in a dramatic decrease in tumor burden and also led to long-term animal survival in every treated animal (10/10 mice remained disease-free at day 100). Importantly, the PEV and cisplatin combination was therapeutically selective and very well tolerated in mice. Our collective data provide a strong rationale for the clinical investigation of NAE inhibition as a novel strategy to augment cisplatin efficacy in patients with head and neck cancer. Citation Format: Trace Jones, Claudia Espitia, Jennifer Carew, Steffan T. Nawrocki. Targeting Neddylation synergistically augments the activity of cisplatin in head and neck cancer models to yield long-term survival by modulating key DNA damage and redox pathways [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 5211.
- Published
- 2020
- Full Text
- View/download PDF
9. Junctional adhesion molecule-A is overexpressed in advanced multiple myeloma and determines response to oncolytic reovirus
- Author
-
Steffan T. Nawrocki, Jennifer S. Carew, Claudia M. Espitia, Weiguo Zhao, Kevin R. Kelly, Fenghuang Zhan, Erik Wendlandt, and Guido Tricot
- Subjects
Adult ,NOXA ,Blotting, Western ,Reoviridae ,Antineoplastic Agents ,Apoptosis ,Receptors, Cell Surface ,Drug resistance ,Biology ,reovirus ,Bortezomib ,Refractory ,Microscopy, Electron, Transmission ,Cell Line, Tumor ,Reolysin ,medicine ,Humans ,Multiple myeloma ,JAM-A ,Aged ,Oncolytic Virotherapy ,Cell adhesion molecule ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Profiling ,Middle Aged ,medicine.disease ,biology.organism_classification ,3. Good health ,Oncolytic virus ,Gene Expression Regulation, Neoplastic ,myeloma ,Oncology ,Drug Resistance, Neoplasm ,Immunology ,Mutation ,Cancer research ,Disease Progression ,ras Proteins ,RNA Interference ,Multiple Myeloma ,Cell Adhesion Molecules ,medicine.drug ,Research Paper - Abstract
Despite the development of several new agents for multiple myeloma (MM) therapy over the last decade, drug resistance continues to be a significant problem. Patients with relapsed/refractory disease have high mortality rates and desperately need new precision approaches that directly target specific molecular features that are prevalent in the refractory setting. Reolysin is a proprietary formulation of reovirus for cancer therapy that has demonstrated efficacy in multiple clinical trials. Its selective effects against solid tumors have been largely attributed to RAS-mediated control of reovirus replication. However, the mechanisms regulating its preferential anti-neoplastic effects in MM and other hematological malignancies have not been rigorously studied. Here we report that the reovirus receptor, junctional adhesion molecule-A (JAM-A) is highly expressed in primary cells from patients with MM and the majority of MM cell lines compared to normal controls. A series of experiments demonstrated that JAM-A expression, rather than RAS, was required for Reolysin-induced cell death in MM models. Notably, analysis of paired primary MM specimens revealed that JAM-A expression was significantly increased at relapse compared to diagnosis. Two different models of acquired resistance to bortezomib also displayed both higher JAM-A expression and elevated sensitivity to Reolysin compared to parental cells, suggesting that Reolysin may be an effective agent for patients with relapsed/refractory disease due to their high JAM-A levels. Taken together, these findings support further investigation of Reolysin for the treatment of patients with relapsed/refractory MM and of JAM-A as a predictive biomarker for sensitivity to Reolysin-induced cell death.
- Published
- 2015
10. Comprehensive quantitative proteomic profiling of the pharmacodynamic changes induced by MLN4924 in acute myeloid leukemia cells establishes rationale for its combination with azacitidine
- Author
-
Jaroslaw P. Maciejewski, Claudia M. Espitia, Anthony Possemato, Hetty E. Carraway, Mikkael A. Sekeres, Steffan T. Nawrocki, Sean A. Beausoleil, Aziz Nazha, Jennifer S. Carew, Yingchun Han, Valeria Visconte, Anjali S. Advani, and Kevin R. Kelly
- Subjects
0301 basic medicine ,Proteomics ,Cancer Research ,azacitidine ,Myeloid ,Azacitidine ,MLN4924 ,Cyclopentanes ,Biology ,acute myeloid leukemia ,Bioinformatics ,Article ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,Cell Line, Tumor ,medicine ,Humans ,skin and connective tissue diseases ,Proteomic Profiling ,Myeloid leukemia ,Hematology ,medicine.disease ,3. Good health ,Neoplasm Proteins ,Leukemia ,Leukemia, Myeloid, Acute ,030104 developmental biology ,medicine.anatomical_structure ,Pyrimidines ,Oncology ,030220 oncology & carcinogenesis ,Pharmacodynamics ,proteome profiling ,Cancer research ,sense organs ,medicine.drug - Abstract
Comprehensive quantitative proteomic profiling of the pharmacodynamic changes induced by MLN4924 in acute myeloid leukemia cells establishes rationale for its combination with azacitidine
- Published
- 2015
11. Combined autophagy and HDAC inhibition
- Author
-
Steffan T. Nawrocki, Claudia M. Espitia, Alain C. Mita, Leslie Wood, Ravi K. Amaravadi, Devalingam Mahalingam, Lisa E. Davis, John Sarantopoulos, Francis J. Giles, Monica M. Mita, Jennifer S. Carew, and Tyler J. Curiel
- Subjects
Adult ,Male ,Maximum Tolerated Dose ,Nausea ,Biology ,Pharmacology ,Hydroxamic Acids ,Pharmacokinetics ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,Autophagy ,medicine ,Humans ,Adverse effect ,Molecular Biology ,Vorinostat ,Aged ,Demography ,Neoplasm Staging ,Aged, 80 and over ,Hydroxychloroquine ,Cell Biology ,Middle Aged ,Neoplasm Proteins ,Histone Deacetylase Inhibitors ,Treatment Outcome ,Pharmacodynamics ,Toxicity ,Female ,Clinical Research Paper ,medicine.symptom ,medicine.drug - Abstract
We previously reported that inhibition of autophagy significantly augmented the anticancer activity of the histone deacetylase (HDAC) inhibitor vorinostat (VOR) through a cathepsin D-mediated mechanism. We thus conducted a first-in-human study to investigate the safety, preliminary efficacy, pharmacokinetics (PK), and pharmacodynamics (PD) of the combination of the autophagy inhibitor hydroxychloroquine (HCQ) and VOR in patients with advanced solid tumors. Of 27 patients treated in the study, 24 were considered fully evaluable for study assessments and toxicity. Patients were treated orally with escalating doses of HCQ daily (QD) (d 2 to 21 of a 21-d cycle) in combination with 400 mg VOR QD (d one to 21). Treatment-related adverse events (AE) included grade 1 to 2 nausea, diarrhea, fatigue, weight loss, anemia, and elevated creatinine. Grade 3 fatigue and/or myelosuppression were observed in a minority of patients. Fatigue and gastrointestinal AE were dose-limiting toxicities. Six-hundred milligrams HCQ and 400 mg VOR was established as the maximum tolerated dose and recommended phase II regimen. One patient with renal cell carcinoma had a confirmed durable partial response and 2 patients with colorectal cancer had prolonged stable disease. The addition of HCQ did not significantly impact the PK profile of VOR. Treatment-related increases in the expression of CDKN1A and CTSD were more pronounced in tumor biopsies than peripheral blood mononuclear cells. Based on the safety and preliminary efficacy of this combination, additional clinical studies are currently being planned to further investigate autophagy inhibition as a new approach to increase the efficacy of HDAC inhibitors.
- Published
- 2014
- Full Text
- View/download PDF
12. Disrupting Protein NEDDylation with MLN4924 Is a Novel Strategy to Target Cisplatin Resistance in Ovarian Cancer
- Author
-
Sean A. Beausoleil, Michael Milhollen, Stephen J. Blakemore, Anthony Possemato, Claudia M. Espitia, Pete Smith, Michael P. Thomas, Allison Berger, Steffan T. Nawrocki, Kevin R. Kelly, and Jennifer S. Carew
- Subjects
Cancer Research ,Proteome ,Cell Survival ,DNA damage ,Apoptosis ,Cyclopentanes ,Biology ,Pharmacology ,medicine.disease_cause ,Mitochondrial Proteins ,Mice ,Protein neddylation ,Ovarian tumor ,medicine ,Animals ,Humans ,Tumor Stem Cell Assay ,Ovarian Neoplasms ,Cisplatin ,Gene knockdown ,NF-kappa B ,Membrane Proteins ,medicine.disease ,Xenograft Model Antitumor Assays ,Tumor Burden ,Pyrimidines ,Oncology ,Drug Resistance, Neoplasm ,Female ,Neddylation ,Apoptosis Regulatory Proteins ,Reactive Oxygen Species ,Ovarian cancer ,Oxidative stress ,DNA Damage ,medicine.drug - Abstract
Purpose: Ovarian cancer has the highest mortality rate of all female reproductive malignancies. Drug resistance is a major cause of treatment failure and novel therapeutic strategies are urgently needed. MLN4924 is a NEDDylation inhibitor currently under investigation in multiple phase I studies. We investigated its anticancer activity in cisplatin-sensitive and -resistant ovarian cancer models. Experimental Design: Cellular sensitivity to MLN4924/cisplatin was determined by measuring viability, clonogenic survival, and apoptosis. The effects of drug treatment on global protein expression, DNA damage, and reactive oxygen species generation were determined. RNA interference established natural born killer/bcl-2–interacting killer (NBK/BIK) as a regulator of therapeutic sensitivity. The in vivo effects of MLN4924/cisplatin on tumor burden and key pharmacodynamics were assessed in cisplatin-sensitive and -resistant xenograft models. Results: MLN4924 possessed significant activity against both cisplatin-sensitive and -resistant ovarian cancer cells and provoked the stabilization of key NEDD8 substrates and regulators of cellular redox status. Notably, MLN4924 significantly augmented the activity of cisplatin against cisplatin-resistant cells, suggesting that aberrant NEDDylation may contribute to drug resistance. MLN4924 and cisplatin cooperated to induce DNA damage, oxidative stress, and increased expression of the BH3-only protein NBK/BIK. Targeted NBK/BIK knockdown diminished the proapoptotic effects of the MLN4924/cisplatin combination. Administration of MLN4924 to mice bearing ovarian tumor xenografts significantly increased the efficacy of cisplatin against both cisplatin-sensitive and -resistant tumors. Conclusions: Our collective data provide a rationale for the clinical investigation of NEDD8-activating enzyme (NAE) inhibition as a novel strategy to augment cisplatin efficacy in patients with ovarian cancer and other malignancies. Clin Cancer Res; 19(13); 3577–90. ©2013 AACR.
- Published
- 2013
- Full Text
- View/download PDF
13. Reovirus therapy stimulates endoplasmic reticular stress, NOXA induction, and augments bortezomib-mediated apoptosis in multiple myeloma
- Author
-
Babatunde O. Oyajobi, Steffan T. Nawrocki, Claudia M. Espitia, Kevin R. Kelly, Matthew C. Coffey, Devalingam Mahalingam, Jennifer S. Carew, and Francis J. Giles
- Subjects
Cancer Research ,Orthoreovirus, Mammalian ,Transplantation, Heterologous ,Antineoplastic Agents ,Apoptosis ,Biology ,Bortezomib ,Small hairpin RNA ,Mice ,hemic and lymphatic diseases ,Reolysin ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,Cells, Cultured ,Oncolytic Virotherapy ,Gene knockdown ,Endoplasmic reticulum ,Endoplasmic Reticulum Stress ,Boronic Acids ,Oncolytic virus ,Proto-Oncogene Proteins c-bcl-2 ,Pyrazines ,Immunology ,Cancer research ,Unfolded protein response ,Multiple Myeloma ,Neoplasm Transplantation ,medicine.drug - Abstract
Oncolytic virotherapy with reovirus has demonstrated anti-cancer activity and minimal toxicity in clinical trials, but the mechanisms underlying these effects have not been fully elucidated. Reolysin, a proprietary formulation of reovirus for cancer therapy, stimulated selective viral replication and apoptosis in multiple myeloma (MM) cells. Reolysin-mediated apoptosis was associated with an induction of endoplasmic reticular (ER) stress-related gene expression, swelling of the endoplasmic reticulum, increases in intracellular calcium levels and a strong induction of the Bcl-2 homology 3 (BH3)-only pro-apoptotic protein NOXA. Knockdown of NOXA expression by short hairpin RNA significantly reduced the pro-apoptotic effects of Reolysin. We next showed that co-administration of Reolysin and bortezomib resulted in the dual accumulation of viral and ubiquitinated proteins, which led to enhanced ER stress, NOXA induction and apoptosis. Importantly, the combination of reovirus infection and proteasomal inhibition significantly decreased tumor burden in a xenograft and syngeneic bone disease model of MM without exhibiting adverse side effects. Our study establishes ER stress stimulation and NOXA induction as novel mediators of reovirus-induced apoptosis. Furthermore, reovirus infection can be used as a promising approach to augment the anti-myeloma activity of bortezomib by promoting additional stress to the endoplasmic reticulum of MM cells.
- Published
- 2011
- Full Text
- View/download PDF
14. Targeting PIM kinase activity significantly augments the efficacy of cytarabine
- Author
-
Pietro Taverna, Jennifer S. Carew, Steffan T. Nawrocki, Kevin R. Kelly, Francis J. Giles, Gavin Choy, Claudia M. Espitia, and Swaminathan Padmanabhan
- Subjects
business.industry ,Cytarabine ,medicine ,Hematology ,Drug resistance ,Pharmacology ,Kinase activity ,business ,medicine.drug - Published
- 2011
- Full Text
- View/download PDF
15. Oncolytic Reovirus Is an Effective Treatment for Histone Deacetylase Inhibitor Resistant T-Cell Lymphoma
- Author
-
Jennifer S. Carew, Ning Qu, Steffan T. Nawrocki, Daniel O. Persky, Claudia M. Espitia, and Shariful Islam
- Subjects
business.industry ,medicine.drug_class ,Immunology ,Histone deacetylase inhibitor ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Oncolytic virus ,Romidepsin ,chemistry.chemical_compound ,chemistry ,Panobinostat ,Cancer research ,Medicine ,T-cell lymphoma ,Histone deacetylase ,business ,Belinostat ,Vorinostat ,medicine.drug - Abstract
Aberrant gene expression plays a pivotal role during tumorigenesis and cancer progression. The acetylation status of histones is an important determinant of gene expression and is controlled by two opposing classes of enzymes: histones acetyl transferases (HATs) and histone deacetylases (HDACs). The deacetylation of histones is associated with repression of key tumor suppressor genes and has been linked to HDAC overexpression in multiple forms of cancer including lymphomas. Several HDAC inhibitors have been FDA approved for T-cell lymphoma (TCL) therapy including belinostat, vorinostat, and romidepsin. Despite the promising anti-lymphoma activity of HDAC inhibitors as a drug class, resistance is a significant clinical issue. Identification of new strategies that are more effective in the drug resistant patient population is a high priority, but the mechanisms underlying HDAC inhibitor-induced cell death and the development of drug resistance are not completely understood. Elucidating the molecular mechanisms driving HDAC inhibitor resistance and/or the specific targets that are altered in drug-resistant cells may facilitate the development of strategies that overcome drug resistance and are effective for refractory patients. To pursue this goal, we generated novel TCL cell line models of acquired HDAC inhibitor resistance through repeated exposure to belinostat. The sensitivity of parental and resistant TCL cells to belinostat and other clinically relevant HDAC inhibitors was initially characterized using cell viability and flow cytometric assays. Notably, belinostat-resistant cells displayed significant cross-resistance to other HDAC inhibitors including vorinostat, romidepsin, panobinostat, and ricolinostat. This indicates that TCL patients that fail one HDAC inhibitor regimen may not benefit significantly from subsequent treatment with other drugs of this class. Consistent with a lack of sensitivity to HDAC inhibitors, the resistant cells failed to induce increased acetylated histones, tubulin acetylation (an HDAC6 target), and exhibited reduced upregulation of the CDK inhibitor p21 following belinostat treatment. In agreement with the absence of these hallmark characteristics of HDAC inhibition, belinostat also failed to cleave caspase-3 in the belinostat-resistant cells. Comprehensive transcriptome analysis was conducted to further characterize these new drug-resistant models and identify potential mechanisms of resistance and targets for second-line treatment. Drug resistant cells featured significantly increased basal levels of the reovirus receptor junctional adhesion molecule-A (JAM-A) as well as decreased JAK/STAT activity, a key antiviral response pathway. Based on these findings, we investigated the efficacy of the proprietary clinical formulation of reovirus (Reolysin) in parental and drug-resistant models. Our investigation revealed that belinostat-resistant cells displayed enhanced sensitivity to oncolytic reovirus-induced cell death compared to their parental counterparts. The increased benefit of Reolysin in resistant models was linked to elevated viral loads and more efficient endoplasmic reticular stress-mediated apoptosis. The heightened sensitivity of HDAC-resistant TCL cells to Reolysin was further validated in parental and belinostat-resistant T-cell lymphoma xenograft models. Collectively, these data demonstrate that oncolytic reovirus may be a novel therapeutic approach to treat T-cell lymphoma patients that are relapsed/refractory to HDAC inhibitors. We are currently planning an early phase clinical trial to further test the safety and benefit of this new approach. Disclosures Persky: Morphosys (IDMC): Consultancy; Merck: Research Funding; Spectrum: Research Funding; Genentech: Honoraria.
- Published
- 2018
- Full Text
- View/download PDF
16. Abstract NTOC-102: TARGETING NEDDYLATION TO OVERCOME CISPLATIN RESISTANCE IN OVARIAN CANCER
- Author
-
Claudia M. Espitia, Jennifer S. Carew, Steffan T. Nawrocki, and Kevin R. Kelly
- Subjects
Cisplatin ,Cancer Research ,Chemotherapy ,medicine.medical_treatment ,Cancer ,Drug resistance ,Biology ,Pharmacology ,medicine.disease ,NEDD8 ,Ovarian tumor ,Oncology ,medicine ,Cancer research ,Neddylation ,Ovarian cancer ,medicine.drug - Abstract
PURPOSE: Ovarian cancer has the highest mortality rate of all female reproductive malignancies. Drug resistance is a major problem that limits overall survival and novel therapeutic strategies are urgently needed. The inappropriate loss of several NEDD8-cullin-RING ubiquitin ligase (CRL) targets has been linked to cancer progression and drug resistance, suggesting that this pathway may be a promising new therapeutic target. MLN4924 is a first-in-class NEDD8 activating enzyme (NAE) inhibitor currently under investigation in multiple clinical studies. We investigated its anticancer activity and pharmacodynamic effects in cisplatin-sensitive and cisplatin-resistant ovarian cancer models. EXPERIMENTAL PROCEDURES: Cellular sensitivity to MLN4924/cisplatin was determined by measuring viability, senescence, and apoptosis. The effects of drug treatment on global protein expression, DNA damage, and reactive oxygen species generation were determined. RNA interference approaches were used to identify and validate regulators of therapeutic sensitivity. The in vivo effects of MLN4924/cisplatin on tumor burden and key pharmacodynamic endpoints were assessed in cisplatin-sensitive and -resistant xenograft models. RESULTS: Intrinsic resistance to cisplatin did not significantly impact the efficacy of MLN4924 against ovarian cancer cells, indicating that this agent may be useful for patients with platinum-resistant disease. Detailed pharmacodynamic analyses showed that treatment with MLN4924 induced the stabilization of key NEDD8 substrates and regulators of cellular redox status. Notably, cisplatin-resistant cells displayed significantly higher basal levels of NEDDylated cullins and NEDDylation activity than cisplatin-sensitive cells. Further investigation demonstrated that MLN4924 significantly augmented the activity of cisplatin against cisplatin-resistant cells. This suggests that aberrant NEDDylation may contribute to drug resistance and that targeting this pathway may be a promising strategy to restore chemosensitivity. MLN4924 and cisplatin cooperated to induce DNA damage, oxidative stress, and increased expression of the BH3-only protein NBK/BIK. Targeted NBK/BIK knockdown diminished the pro-apoptotic effects of the MLN4924/cisplatin combination. In addition, administration of MLN4924 to mice bearing ovarian tumor xenografts significantly increased the efficacy of cisplatin against both cisplatin-sensitive and cisplatin-resistant tumors. CONCLUSIONS: Our collective data provide a rationale for the clinical investigation of NAE inhibition as a novel strategy to augment cisplatin efficacy in patients with ovarian cancer and other malignancies where platinum-based chemotherapy is utilized. Citation Format: Steffan T. Nawrocki, Kevin R. Kelly, Claudia M. Espitia, and Jennifer S. Carew. TARGETING NEDDYLATION TO OVERCOME CISPLATIN RESISTANCE IN OVARIAN CANCER [abstract]. In: Proceedings of the 11th Biennial Ovarian Cancer Research Symposium; Sep 12-13, 2016; Seattle, WA. Philadelphia (PA): AACR; Clin Cancer Res 2017;23(11 Suppl):Abstract nr NTOC-102.
- Published
- 2017
- Full Text
- View/download PDF
17. Targeting Survivin Inhibits Renal Cell Carcinoma Progression and Enhances the Activity of Temsirolimus
- Author
-
Alain C. Mita, Weiguo Zhao, Claudia M. Espitia, Jennifer S. Carew, Steffan T. Nawrocki, and Monica M. Mita
- Subjects
Cancer Research ,Cell Survival ,Survivin ,Immunoblotting ,Mice, Nude ,Apoptosis ,Biology ,urologic and male genital diseases ,medicine.disease_cause ,Inhibitor of Apoptosis Proteins ,Renal cell carcinoma ,Proliferating Cell Nuclear Antigen ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Gene silencing ,Animals ,neoplasms ,Carcinoma, Renal Cell ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Sirolimus ,Mice, Inbred BALB C ,Reverse Transcriptase Polymerase Chain Reaction ,Imidazoles ,Drug Synergism ,medicine.disease ,Immunohistochemistry ,Xenograft Model Antitumor Assays ,female genital diseases and pregnancy complications ,Temsirolimus ,Kidney Neoplasms ,Gene Expression Regulation, Neoplastic ,Oncology ,Tumor progression ,Cancer cell ,Cancer research ,Disease Progression ,Female ,RNA Interference ,Carcinogenesis ,medicine.drug ,Naphthoquinones - Abstract
Elevated expression of the antiapoptotic factor survivin has been implicated in cancer cell survival and disease progression. However, its specific contribution to renal cell carcinoma (RCC) pathogenesis is not well defined. We investigated the roles of survivin in RCC tumor progression, resistance to mTOR inhibitors, and evaluated the therapeutic activity of the survivin suppressant YM155 in RCC models. Here, we report that survivin expression levels were significantly higher in RCC cell lines compared with normal renal cells. Stable targeted knockdown of survivin completely abrogated the ability of 786-O RCC tumors to grow in mice, thus demonstrating its importance as a regulator of RCC tumorigenesis. We next explored multiple strategies to therapeutically inhibit survivin function in RCC. Treatment with the mTOR inhibitor temsirolimus partially diminished survivin levels and this effect was augmented by the addition of YM155. Further analyses revealed that, in accordance with their combined anti-survivin effects, YM155 significantly improved the anticancer activity of temsirolimus in a panel of RCC cell lines in vitro and in xenograft models in vivo. Similar to pharmacologic inhibition of survivin, shRNA-mediated silencing of survivin expression not only inhibited RCC tumor growth, but also significantly sensitized RCC cells to temsirolimus therapy. Subsequent experiments demonstrated that the effectiveness of this dual survivin/mTOR inhibition strategy was mediated by a potent decrease in survivin levels and corresponding induction of apoptosis. Our findings establish survivin inhibition as a novel approach to improve RCC therapy that warrants further investigation. Mol Cancer Ther; 14(6); 1404–13. ©2015 AACR.
- Published
- 2014
18. A Novel Oncolytic Reovirus Immune Priming Strategy Dramatically Enhances the Efficacy of Anti-PD-L1 Antibody Therapy Against Multiple Myeloma
- Author
-
Valeria Visconte, Weiguo Zhao, Kevin R. Kelly, Steffan T. Nawrocki, Jennifer S. Carew, Matthew C. Coffey, and Claudia M. Espitia
- Subjects
biology ,Bortezomib ,business.industry ,medicine.medical_treatment ,Immunology ,Cell Biology ,Hematology ,Biochemistry ,Immune checkpoint ,Targeted therapy ,Oncolytic virus ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,030220 oncology & carcinogenesis ,Reolysin ,Cancer cell ,biology.protein ,medicine ,Cancer research ,Antibody ,business ,030215 immunology ,medicine.drug - Abstract
Multiple myeloma (MM) is the second most common adult hematologic malignancy in the United States and is characterized by a unique form of progressive bone destruction. Despite the development of new treatments such as proteasome inhibitors and immunomodulatory agents, patients with high-risk disease have a median survival of only 2-3 years, highlighting the demand for more effective therapeutic strategies. Oncolytic viral formulations represent a promising new class of anticancer agents that may have important applications in precision medicine. Recent studies have demonstrated that reoviruses specifically replicate only in cancer cells and this led to the development of a reovirus-based oncolytic viral therapy called Reolysin. We have demonstrated that reoviruses selectively replicate in MM cells and Reolysin possesses significant activity in preclinical in vitro and in vivo MM models. These findings established the framework for an ongoing investigator-initiated phase 1b clinical study of Reolysin in combination with bortezomib and dexamethasone in patients with relapsed/refractory MM. Recent gene ontology analyses of RPMI-8226 and U266 MM cells treated with Reolysin revealed that reovirus exposure triggers a highly significant transient increase in CD274(programmed death 1 ligand, PD-L1) in MM cell lines. Reolysin-mediated PD-L1 upregulation was confirmed by immunoblotting, qRT-PCR, and flow cytometric analyses in MM cell lines and primary patient specimens treated with Reolysin. Increased PD-L1 expression was also detected by immunohistochemistry in MM tumor samples collected from mice treated with Reolysin. Adaptive resistance mediated by inhibitory ligands such as PD-L1 has emerged as an important mechanism of malignant cell survival and has led to the development of new agents that disrupt the PD-L1/PD-1 immune checkpoint. These agents have exhibited dramatic efficacy in certain forms of cancer including melanoma and lung cancer. Analysis of specimens from patients treated on clinical trials with these agents indicates that high basal expression of PD-L1 on tumor cells may be necessary to elicit significant clinical benefit. Notably, most MM cell lines and primary CD138+ cells from MM patients do not overexpress PD-L1 compared to normal plasma cells and this may preclude patients with MM from optimally benefiting from immune checkpoint therapy. However, novel immune priming strategies that stimulate transient upregulation of PD-L1 on malignant cells could potentially render agents that target the PD-L1/PD-1 axis significantly more effective for a broader range of malignancies including MM. We hypothesized that Reolysin could be used as a precision immune priming agent to potentiate the anti-MM efficacy of PD-L1 targeted therapy by rendering MM cells vulnerable to PD-L1 inhibition through the transient upregulation of target expression. To investigate this therapeutic approach, 5TGM1-luc murine MM cells were injected IV into immunocompetent mice to generate MM bone disease. After disease was established, mice were randomized into groups and treated with vehicle, Reolysin (5 x 108 TCID50, Q7D), murine anti-PD-L1 antibody (200 mg/mouse, Q2D) or the combination for 5 weeks. Mice treated with the combination demonstrated decreased disease burden as measured by bioluminescent imaging and also showed reduced IgG2bk levels (specific IgG secreted by 5TGM1 cells) by ELISA. Importantly, the combination also led to increased overall animal survival compared to vehicle control and either single agent treatment (P Disclosures Kelly: Novartis: Consultancy, Speakers Bureau; Pharmacyclics: Consultancy, Speakers Bureau; Amgen: Consultancy, Membership on an entity's Board of Directors or advisory committees. Coffey:Oncolytics Biotech: Employment.
- Published
- 2016
- Full Text
- View/download PDF
19. Targeting Aurora A kinase activity with the investigational agent alisertib increases the efficacy of cytarabine through a FOXO-dependent mechanism
- Author
-
Swaminathan Padmanabhan, Johnny J. Yang, Jennifer S. Carew, Claudia M. Espitia, Mengkun Zhang, Steffan T. Nawrocki, Francis J. Giles, Kevin R. Kelly, and Jeffrey Ecsedy
- Subjects
Cancer Research ,Cell Survival ,Aurora A kinase ,Apoptosis ,HL-60 Cells ,Mice, SCID ,Pharmacology ,Protein Serine-Threonine Kinases ,Article ,chemistry.chemical_compound ,Mice ,Aurora Kinases ,hemic and lymphatic diseases ,Cell Line, Tumor ,Proto-Oncogene Proteins ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,Viability assay ,Molecular Targeted Therapy ,Protein Kinase Inhibitors ,Aurora Kinase A ,Gene knockdown ,Bcl-2-Like Protein 11 ,business.industry ,Cell Cycle ,Forkhead Box Protein O3 ,Cytarabine ,Myeloid leukemia ,Membrane Proteins ,Drug Synergism ,Forkhead Transcription Factors ,Azepines ,Cell cycle ,Xenograft Model Antitumor Assays ,Leukemia, Myeloid, Acute ,Pyrimidines ,Oncology ,chemistry ,Alisertib ,Female ,business ,Apoptosis Regulatory Proteins ,Cyclin-Dependent Kinase Inhibitor p27 ,medicine.drug - Abstract
Novel therapies are urgently needed to improve clinical outcomes for patients with acute myeloid leukemia (AML). The investigational drug alisertib (MLN8237) is a novel Aurora A kinase inhibitor being studied in multiple Phase I and II studies. We investigated the preclinical efficacy and pharmacodynamics of alisertib in AML cell lines, primary AML cells, and mouse models of AML. Here we report that alisertib disrupted cell viability, diminished clonogenic survival, induced expression of the FOXO3a targets p27 and BIM, and triggered apoptosis. A link between Aurora A expression and sensitivity to ara-C was established, suggesting that Aurora A inhibition may be a promising strategy to increase the efficacy of ara-C. Accordingly, alisertib significantly potentiated the anti-leukemic activity of ara-C in both AML cell lines and primary blasts. Targeted FOXO3a knockdown significantly blunted the pro-apoptotic effects of the alisertib/ara-C combination, indicating that it is an important regulator of sensitivity to these agents. In vivo studies demonstrated that alisertib significantly augmented the efficacy of ara-C without affecting its pharmacokinetic profile and led to the induction of p27 and BIM. Our collective data indicate that targeting Aurora A with alisertib represents a novel approach to increase the efficacy of ara-C that warrants further investigation.
- Published
- 2012
20. Lucanthone Is a Novel Inhibitor of Autophagy That Induces Cathepsin D-mediated Apoptosis*
- Author
-
Claudia M. Espitia, Guru Reddy, Francis J. Giles, Jennifer S. Carew, Steffan T. Nawrocki, Juan A. Esquivel, Kevin R. Kelly, and Devalingam Mahalingam
- Subjects
Programmed cell death ,medicine.drug_class ,Cathepsin D ,Antineoplastic Agents ,Apoptosis ,Breast Neoplasms ,Biology ,Hydroxamic Acids ,Biochemistry ,Permeability ,chemistry.chemical_compound ,Schistosomicides ,Cell Line, Tumor ,Phagosomes ,Sequestosome-1 Protein ,medicine ,Autophagy ,Humans ,Molecular Biology ,Vorinostat ,Adaptor Proteins, Signal Transducing ,Oligonucleotide Array Sequence Analysis ,Lucanthone ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Profiling ,Histone deacetylase inhibitor ,Drug Synergism ,Cell Biology ,Intracellular Membranes ,Cell biology ,chemistry ,Cancer cell ,Tumor Suppressor Protein p53 ,Lysosomes ,Microtubule-Associated Proteins ,medicine.drug ,Signal Transduction - Abstract
Cellular stress induced by nutrient deprivation, hypoxia, and exposure to many chemotherapeutic agents activates an evolutionarily conserved cell survival pathway termed autophagy. This pathway enables cancer cells to undergo self-digestion to generate ATP and other essential biosynthetic molecules to temporarily avoid cell death. Therefore, disruption of autophagy may sensitize cancer cells to cell death and augment chemotherapy-induced apoptosis. Chloroquine and its analog hydroxychloroquine are the only clinically relevant autophagy inhibitors. Because both of these agents induce ocular toxicity, novel inhibitors of autophagy with a better therapeutic index are needed. Here we demonstrate that the small molecule lucanthone inhibits autophagy, induces lysosomal membrane permeabilization, and possesses significantly more potent activity in breast cancer models compared with chloroquine. Exposure to lucanthone resulted in processing and recruitment of microtubule-associated protein 1 light chain 3 (LC3) to autophagosomes, but impaired autophagic degradation as revealed by transmission electron microscopy and the accumulation of p62/SQSTM1. Microarray analysis, qRT-PCR, and immunoblotting determined that lucanthone stimulated a large induction in cathepsin D, which correlated with cell death. Accordingly, knockdown of cathepsin D reduced lucanthone-mediated apoptosis. Subsequent studies using p53(+/+) and p53(-/-) HCT116 cells established that lucanthone induced cathepsin D expression and reduced cancer cell viability independently of p53 status. In addition, lucanthone enhanced the anticancer activity of the histone deacetylase inhibitor vorinostat. Collectively, our results demonstrate that lucanthone is a novel autophagic inhibitor that induces apoptosis via cathepsin D accumulation and enhances vorinostat-mediated cell death in breast cancer models.
- Published
- 2010
21. Vorinostat enhances the activity of temsirolimus in renal cell carcinoma through suppression of survivin levels
- Author
-
Kelli Oberheu, Alain C. Mita, Kevin R. Kelly, Steffan T. Nawrocki, Ernest Medina, Jennifer S. Carew, Claudia M. Espitia, Sabrina Smith, Ronan T. Swords, Juan A. Esquivel, Devalingam Mahalingam, Francis J. Giles, and Monica M. Mita
- Subjects
Cancer Research ,Angiogenesis ,medicine.drug_class ,Survivin ,Mice, Nude ,Angiogenesis Inhibitors ,Apoptosis ,Biology ,Hydroxamic Acids ,Inhibitor of Apoptosis Proteins ,Mice ,In vivo ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,neoplasms ,Vorinostat ,Carcinoma, Renal Cell ,PI3K/AKT/mTOR pathway ,Sirolimus ,Histone deacetylase inhibitor ,Drug Synergism ,Xenograft Model Antitumor Assays ,Temsirolimus ,Kidney Neoplasms ,Repressor Proteins ,Oncology ,Cancer research ,Histone deacetylase ,Microtubule-Associated Proteins ,medicine.drug - Abstract
Purpose: The mammalian target of rapamycin (mTOR) inhibitor temsirolimus has exhibited promising anticancer activity for the treatment of renal cell cancers (RCC). Survivin expression has been implicated in drug resistance and reducing its levels with the histone deacetylase (HDAC) inhibitor vorinostat may enhance the anticancer activity of temsirolimus. Experimental Design: The sensitivity of RCC cell lines to the combination of temsirolimus and vorinostat was determined by measuring cell viability, clonogenic survival, and apoptosis. The effects of this combination on survivin levels were determined in vitro and in vivo. Survivin expression was silenced using small interfering RNA to evaluate its role in determining sensitivity to temsirolimus and vorinostat. The effect of the combination on angiogenesis was also determined in RCC xenograft models. Results: Vorinostat synergistically improved the anticancer activity of temsirolimus in a panel of RCC cell lines in vitro and in two xenograft models in vivo. While each single agent led to a modest decrease in survivin levels, the combination dramatically reduced its expression, which correlated with an induction of apoptosis. Silencing survivin levels induced apoptosis and significantly improved the efficacy of temsirolimus and vorinostat. In addition, the temsirolimus/vorinostat combination led to a strong reduction in angiogenesis. Conclusions: Vorinostat augmented the anticancer activity of temsirolimus in both in vitro and in vivo models of RCC. The effectiveness of the combination was due to a decrease in survivin levels and corresponding induction of apoptosis, and enhanced inhibition of angiogenesis. Targeting survivin may be a promising therapeutic strategy to improve RCC therapy. Clin Cancer Res; 16(1); 141–53
- Published
- 2009
22. Antagonizing HDAC6 Activity: A New Strategy to Maximize the Anti-Myeloma Effects of BET Inhibition
- Author
-
Weiguo Zhao, Kevin R. Kelly, Steffan T. Nawrocki, Jennifer S. Carew, Valeria Visconte, and Claudia M. Espitia
- Subjects
BRD4 ,Gene knockdown ,Oncogene ,Cell growth ,Bortezomib ,Immunology ,Cell Biology ,Hematology ,HDAC6 ,Biology ,Biochemistry ,Apoptosis ,medicine ,Cancer research ,Vorinostat ,medicine.drug - Abstract
Multiple myeloma (MM) is an incurable plasma cell malignancy and represents the second most common adult hematologic malignancy in the United States. MM is relatively asymptomatic during its early stages and as a result, the majority of patients have advanced disease at diagnosis. Innovations in the treatment of MM, including the development of proteasome inhibitors such as bortezomib (Velcade) have improved clinical outcomes. However, many patients fail to respond to these agents or relapse after initial response highlighting the need for novel therapeutic strategies. Constitutive activation of the MYC oncogene is a frequent pathogenic event in MM that drives disease progression. Aberrant MYC transcriptional activity can increase the levels of a number of factors that are associated with disease progression and drug resistance making it an appealing therapeutic target. Recent studies have demonstrated that inhibition of bromodomain and extra terminal (BET) protein family members including BRD4 decreases the expression of c-MYC and other key oncogenic factors. Here, we demonstrate that shRNA-mediated knockdown of BRD4 or treatment with the BET antagonist JQ1 decreased the expression of c-MYC, BCL-xL, and BCL-2, induced p21 levels, diminished cell viability, and triggered apoptosis in MM cell lines. Comprehensive gene expression profiling of the pharmacodynamic effects of JQ1 revealed that the histone deacetylase HDAC6 was also highly significantly elevated in all MM cell lines and primary patient specimens treated with this agent. Several earlier studies demonstrated that aberrant HDAC6 expression/activity contributes to malignant progression and resistance to a number of classes of anticancer agents including proteasome inhibitors. Based on the roles of HDAC6 in malignant pathogenesis, we hypothesized that its induction may reduce the anti-myeloma activity of JQ1. To test this hypothesis, we utilized both genetic and pharmacological approaches to impair HDAC6 function [shRNA-mediated knockdown of HDAC6, the pan-HDAC inhibitor vorinostat, and the HDAC6-selective inhibitor ACY-1215 (rocilinostat)] and evaluated the consequential impact on the anti-MM effects of JQ1. Notably, antagonzing HDAC6 activity synergistically enhanced the activity of JQ1 in a panel of MM cell lines. These effects were also observed in primary CD138+ cells obtained from patients with MM in a manner that was not affected by prior treatment history. The increased efficacy of these therapeutic combinations was associated with further reductions in c-MYC, BCL-2, and BCL-xL along with significant increases in apoptosis induction as evidenced by enhanced caspase-3 cleavage and DNA fragmentation. Importantly, administration of ACY-1215 was very well tolerated (less than 5% mean transient reduction in body weight) and significantly augmented the in vivo anti-myeloma activity of JQ1 in the RPMI-8226 MM xenograft model as disease burden in combination treated animals was substantially lower than those that received either monotherapy. Immunohistochemical analyses demonstrated that the combination of JQ1 and ACY-1215 led to significantly lower MM cell proliferation (PCNA), increased apoptosis (active caspase-3), and diminished expression of c-MYC and BCL-2. These data suggest for the first time that induction of HDAC6 may represent a key mechanism that promotes drug resistance and limits the efficacy of bromodomain inhibitor therapy. Taken together, our findings demonstrate that abrogation of HDAC6 activity with ACY-1215 or vorinostat is a novel approach to augment the efficacy of bromodomain inhibitors in MM that warrants further investigation. Disclosures Carew: Boehringer Ingelheim: Research Funding.
- Published
- 2015
- Full Text
- View/download PDF
23. Small Molecule Inhibition of SIRT Activity: A Novel Therapeutic Approach for Refractory Multiple Myeloma
- Author
-
Yingchun Han, Valeria Visconte, Jennifer S. Carew, Kevin R. Kelly, Steffan T. Nawrocki, and Claudia M. Espitia
- Subjects
Gene knockdown ,biology ,Bortezomib ,Cyclin D ,Immunology ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Carfilzomib ,Pathogenesis ,chemistry.chemical_compound ,Cyclin D1 ,chemistry ,Sirtuin ,biology.protein ,medicine ,Cancer research ,Multiple myeloma ,medicine.drug - Abstract
Multiple myeloma (MM) is the second most common adult hematologic malignancy in the United States and is characterized by a unique form of progressive bone destruction. Although new treatments such as bortezomib (BZ), carfilzomib and IMIDs have provided benefit to many patients with MM, drug resistance remains a very significant problem. Patients diagnosed with high-risk MM in particular are in urgent need of more effective therapeutic strategies in order to improve their survivorship. The sirtuin deacetylases (SIRTs, SIRT1-7) are critical epigenetic regulators that control longevity, cell growth, tumor suppression, and apoptosis. Elevated SIRT expression has been reported in several types of cancer and may promote pathogenesis and drug resistance by increasing the lifespan and survival capacity of malignant cells. Our preliminary analysis of SIRT expression indicated that SIRT1 was consistently expressed at significantly higher levels in primary MM cells from patients with high-risk disease than those with favorable risk MM or normal CD138+ controls. We investigated the role of SIRT1 as a potential regulator of MM pathogenesis and evaluated the benefit of SIRT inhibition as a novel strategy for MM therapy. A series of experiments established a mechanistic relationship between MYC and SIRT1 in that SIRT1 increased MYC protein stability and transcriptional activity. Disruption of SIRT1 function through targeted knockdown or with the small molecule inhibitor tenovin-6 (T6) directly led to the reduction in the expression of key MYC transcriptional targets that play important roles in MM pathogenesis including cyclin D and MAF. Subsequent analyses revealed that U266 and RPMI-8226 cells with acquired BZ resistance exhibited significantly higher basal SIRT1 expression levels (p Disclosures Carew: Boehringer Ingelheim: Research Funding.
- Published
- 2015
- Full Text
- View/download PDF
24. Targeting HDAC6 Is a Novel Approach to Augment the Therapeutic Benefit of Bromodomain Inhibition in Multiple Myeloma
- Author
-
Steffan T. Nawrocki, Claudia M. Espitia, Weiguo Zhao, Kevin R. Kelly, and Jennifer S. Carew
- Subjects
BRD4 ,Gene knockdown ,Oncogene ,Immunology ,Cell Biology ,Hematology ,HDAC6 ,Biology ,Biochemistry ,Bromodomain ,Cancer research ,medicine ,Viability assay ,Histone deacetylase ,Vorinostat ,medicine.drug - Abstract
Multiple myeloma (MM) is a plasma cell disease that represents the second most common adult hematologic malignancy in the United States. Advances in the treatment of MM, including the development of proteasome inhibitors have improved clinical outcomes. However, many patients fail to respond to these newer agents or relapse after initial response. Constitutive activation of the MYC oncogene is a frequent pathogenic event in MM that drives disease progression making it an ideal therapeutic target. Recent studies suggest that inhibition of bromodomain and extra terminal (BET) protein family members including BRD4 decreases the expression of c-MYC and other key oncogenic factors such as BCL-2 and displays significant activity against MM. Here, we demonstrate that shRNA-mediated knockdown of BRD4 or treatment with the BRD4 antagonist JQ1 decreased the expression of c-MYC, BCL-2, and BCL-XL, diminished cell viability, disrupted clonogenic survival, and triggered apoptosis in a panel of MM cell lines. Subsequent assays conducted in MM cell lines confirmed a strong reduction in MYC levels and activity following treatment with JQ1 along with consistent increases in the expression of genes regulated by BRD4 including CDKN1A and HEXIM1. Interestingly, gene expression profiling assays revealed that the histone deacetylase HDAC6 was also highly significantly elevated in all MM cell lines and primary patient specimens treated with JQ1. Based on its ability to alter cellular survival capacity, we hypothesized that HDAC6 induction may reduce the anti-myeloma activity of JQ1. To test this hypothesis, we utilized shRNA-mediated knockdown of HDAC6, the pan-HDAC inhibitor vorinostat, and the HDAC6-selective inhibitor ACY-1215 (rocilinostat) and evaluated the impact of each on the anti-MM effects of JQ1. Abrogation of HDAC6 activity synergistically enhanced the activity of JQ1 in a panel of MM cell lines. These effects were also observed in primary CD138+ cells obtained from patients with MM in a manner that was not affected by prior treatment history. The increased efficacy of these therapeutic combinations was associated with apoptosis induction as evidenced by enhanced caspase-3 cleavage and further reductions in c-MYC expression. These data suggest that HDAC6 induction may represent a key mechanism that promotes drug resistance and/or limits the efficacy of bromodomain inhibitor therapy. Our collective findings indicate that abrogation of HDAC6 activity with ACY-1215 or vorinostat is a novel and promising approach to augment the efficacy of bromodomain inhibitors for MM that warrants further investigation. Disclosures No relevant conflicts of interest to declare.
- Published
- 2014
- Full Text
- View/download PDF
25. The Novel Plk Inhibitor Volasertib Overcomes Cytarabine Resistance in Acute Myeloid Leukemia
- Author
-
Hetty E. Carraway, Nawrocki T. Steffan, Shuang Fu, Mikkael A. Sekeres, Kevin R. Kelly, Anjali S. Advani, Claudia M. Espitia, Yingchun Han, Jaroslaw P. Maciejewski, and Jennifer S. Carew
- Subjects
Immunology ,Myeloid leukemia ,Volasertib ,Cell Biology ,Hematology ,Drug resistance ,Pharmacology ,Biology ,Biochemistry ,PLK1 ,chemistry.chemical_compound ,chemistry ,Apoptosis ,Cancer research ,Cytarabine ,medicine ,CXCL10 ,Ex vivo ,medicine.drug - Abstract
Drug resistance is a major cause of treatment failure for patients with acute myeloid leukemia (AML) and novel approaches that circumvent or overcome resistance mechanisms are critically needed. Polo-like kinase 1 (PLK1) is a serine/threonine kinase that plays an important role in regulating cell cycle progression in M phase. It is overexpressed in a broad range of solid and hematological malignancies. Volasertib (BI 6727) is a novel PLK inhibitor that is currently under investigation in a Phase III clinical trial for patients with AML. Despite its reported clinical anti-AML activity, the efficacy and pharmacodynamics (PD) of volasertib have not been rigorously investigated in AML models in the preclinical setting. Here, we studied the activity and PD effects of volasertib in a panel of human AML cell lines and primary patient specimens. Low nanomolar concentrations of volasertib potently disrupted the viability and clonogenic survival of AML cells. Notably, the specific cytogenetic background of individual cell lines did not significantly impact cellular sensitivity to volasertib. Similarly, primary AML blasts from patients demonstrated ex vivo sensitivity to volasertib regardless of their cytogenetic and molecular features or prior treatment history. This in vitro data set is in line with recent Phase II clinical data in AML patients showing efficacy of volasertib across all cytogenetic risk categories. Volasertib triggered caspase-dependent apoptosis in AML cells in a manner that was preceded by disruption of the spindle assembly checkpoint. Notably, analysis of the activity of volasertib in three different paired cytarabine-sensitive and cytarabine-resistant AML cell line models revealed that volasertib retained its ability to diminish viability and stimulate apoptosis in cells with three log-fold increased cytarabine resistance. These findings suggest that volasertib may be effective for patients that have relapsed or are refractory to cytarabine-based regimens. Combination studies showed that volasertib and cytarabine yielded additive effects in cytarabine-sensitive cells. Gene expression profiling studies demonstrated that the chemokine CXCL10 was the most significantly upregulated gene following short-term treatment with low concentrations of volasertib. Subsequent qRT-PCR and ELISA experiments confirmed drug-induced CXCL10 expression and elevated CXCL10 secretion by AML cells. To our knowledge, this is the first report linking PLK1 inhibition to alterations in CXCL10 expression and secretion. We are currently investigating the specific contribution of CXCL10 to the anti-leukemic activity of volasertib and are elucidating the molecular mechanisms that mediate this effect. Collectively, our findings demonstrate that inhibition of PLK1 with volasertib is a promising new strategy for the treatment of AML that is able to overcome cytarabine resistance. Disclosures No relevant conflicts of interest to declare.
- Published
- 2014
- Full Text
- View/download PDF
26. The Investigational Multi-Targeted Kinase Inhibitor TAK-901 Antagonizes Acute Myeloid Leukemia Pathogenesis: Results of Preclinical Studies
- Author
-
Maki Hasegawa, Patrick T. Griffin, Claudia M. Espitia, Takashi Satou, Ron de Jong, Kevin R. Kelly, Steffan T. Nawrocki, Swaminathan Padmanabhan, Jennifer S. Carew, and Angela Romanelli
- Subjects
Kinase ,business.industry ,Immunology ,Myeloid leukemia ,Cell Biology ,Hematology ,Pharmacology ,Biochemistry ,medicine.anatomical_structure ,Apoptosis ,In vivo ,medicine ,Cytarabine ,Cytotoxic T cell ,Bone marrow ,business ,GADD45A ,medicine.drug - Abstract
Abstract 581 The long-term prognosis for the majority of patients diagnosed with acute myeloid leukemia (AML) is very poor due, in part, to pre-existing myelodysplasia, multidrug resistance, and co-existing morbidities that limit therapeutic options. Novel strategies are essential in order to improve clinical outcomes. TAK-901 is an investigational small molecule kinase inhibitor that is currently being evaluated in Phase I trials. In preclinical studies, TAK-901 has demonstrated significant effects against a number of kinases with important roles in cancer including the Aurora kinases, which are key regulators of mitosis and whose overexpression in cancer promotes genetic instability, malignant pathogenesis, and drug resistance. We hypothesized that simultaneously targeting the activity of the Auroras and other oncogenic kinases with TAK-901 would disrupt AML pathogenesis. In order to test our hypothesis, we investigated the efficacy and pharmacodynamic activity of TAK-901 human AML cell lines, primary AML specimens, and an orthotopic bioluminescent disseminated mouse model of AML. TAK-901 potently diminished the viability of a panel of 8 AML cell lines as well as primary cells obtained from patients with AML. Acute exposure to TAK-901 ablated clonogenic survival, triggered the accumulation of polyploid cells, and induced apoptosis. The cytostatic and cytotoxic effects of TAK-901 were associated with significantly increased expression of the cyclin-dependent kinase inhibitor p27, growth arrest and DNA-damage-inducible 45a (GADD45a), and the BH3-only pro-apoptotic protein PUMA. Chromatin immunoprecipitation (ChIP) assays revealed that the elevation in the expression of these genes caused by administration of TAK-901 was due to increased FOXO3a transcriptional activity. The in vivo anti-leukemic activity of TAK-901 was investigated in a disseminated xenograft mouse model of AML established by intravenous injection of luciferase-expressing MV4-11 cells. IVIS Xenogen imaging was utilized to monitor disease burden throughout the study. In this mouse model, administration of TAK-901 was very well-tolerated and significantly more effective than the standard of care drug cytarabine with respect to suppressing disease progression and prolonging overall survival. Analysis of specimens collected from mice demonstrated that TAK-901 inhibited the homing of AML cells to the bone marrow microenvironment and induced AML cell apoptosis in vivo. Our collective findings indicate that TAK-901 is a novel multi-targeted kinase inhibitor that has significant preclinical activity in AML models and warrants further investigation. Disclosures: Satou: Takeda Pharmaceuticals: Employment. Hasegawa:Takeda Pharmaceuticals: Employment. Romanelli:Millennium Pharmaceuticals: Employment. de Jong:Takeda San Diego: Employment. Carew:Millennium Pharmaceuticals: Research Funding.
- Published
- 2011
- Full Text
- View/download PDF
27. Targeting Pim Kinase Activity with the Novel Small Molecule Inhibitor SGI-1776 Significantly Augments the Efficacy of Cytarabine In Models of Acute Myeloid Leukemia
- Author
-
Kelli Oberheu, Ernest Medina, Pietro Taverna, Steffan T. Nawrocki, Jennifer S. Carew, Kevin R. Kelly, Devalingam Mahalingam, Claudia M. Espitia, Gavin S. Choy, and Francis J. Giles
- Subjects
Cell growth ,Kinase ,Immunology ,Myeloid leukemia ,PIM1 ,Cell Biology ,Hematology ,Biology ,Pharmacology ,Biochemistry ,Apoptosis ,hemic and lymphatic diseases ,Cytarabine ,medicine ,Kinase activity ,Signal transduction ,medicine.drug - Abstract
Abstract 3253 Drug resistance is a major cause of treatment failure for patients with acute myeloid leukemia (AML) and novel strategies that circumvent resistance mechanisms are urgently needed. The Pim kinases (PIM1, PIM2, PIM3) are a small family of proto-oncogenes within the CAMK superfamily with essential roles in the regulation of signal transduction cascades that promote cell survival, proliferation, and drug resistance. Accordingly, overexpression of Pim kinases has been reported in a wide range of malignancies including AML. However, the specific role of Pim kinases as regulators of sensitivity to standard agents utilized in AML therapy remains to be elucidated. We hypothesized that inhibiting Pim kinase activity would significantly increase the efficacy of the standard of care drug cytarabine by disrupting signaling events that enable cells to resist cytotoxic stress. In order to investigate a potential role for Pim kinases as mediators of cytarabine resistance, we first evaluated their expression levels in paired HL-60 cell lines that are sensitive and resistant to cytarabine. Our results showed that the levels of Pim-1 are significantly higher in cytarabine-resistant HL-60 cells compared with their sensitive counterparts. Consistent with this observation, treatment of cultured AML cell lines and primary AML blasts with cytarabine led to a dose-dependent increase in Pim-1 expression as assessed by quantitative RT-PCR and immunoblotting. SGI-1776 is novel small molecule inhibitor of Pim kinase activity that has entered Phase I clinical trials. To further test our hypothesis, we investigated the in vitro efficacy of SGI-1776 in a panel of 9 human AML cell lines. SGI-1776 led to a dose-dependent reduction in viability in all cell lines evaluated. Subsequent experiments showed that SGI-1776 impaired clonogenic survival, diminished cell proliferation as determined by BrdU incorporation, and induced apoptotic cell death. These effects were associated with a significant reduction in the phosphorylation of the Pim kinase substrate and apoptotic regulator Bad as well as an increase in the expression of the cyclin-dependent kinase inhibitor, p27. We next investigated whether inhibiting Pim signaling with SGI-1776 could augment the efficacy of cytarabine. Treatment of AML cells with the combination of cytarabine and SGI-1776 led to significantly greater inhibition of clonogenic survival and induction of apoptosis over what was achieved by either single agent. To validate the therapeutic utility of this combination, vehicle, cytarabine, SGI-1776, or cytarabine and SGI-1776 were administered to mice bearing MOLM-13 AML xenografts for 21 days. Treatment with the combination of these two agents was very well tolerated and significantly increased the efficacy of single agent cytarabine therapy. Immunohistochemical analyses of specimens from mice treated with this regimen revealed that SGI-1776 diminished Bad phosphorylation and cooperated with cytarabine in vivo to promote activation of caspase-3 and inhibit tumor cell proliferation as quantified by the expression of proliferating cell nuclear antigen (PCNA). Collectively, our data demonstrate that antagonizing Pim kinase activity represents a new strategy to increase the therapeutic efficacy of cytarabine. A clinical trial evaluating the safety and efficacy of this combination for patients with AML that are refractory to standard treatments is warranted. Disclosures: Taverna: SuperGen, Inc.: Employment. Choy:SuperGen, Inc.: Employment.
- Published
- 2010
- Full Text
- View/download PDF
28. 580 Reolysin induces endoplasmic reticular stress in multiple myeloma and enhances the activity of bortezomib
- Author
-
Francis J. Giles, Claudia M. Espitia, Babatunde O. Oyajobi, J.S. Carew, Kevin R. Kelly, E. Medina, S.T. Nawrocki, Matthew C. Coffey, and Devalingam Mahalingam
- Subjects
Cancer Research ,Oncology ,Bortezomib ,Chemistry ,Endoplasmic reticulum ,Reolysin ,Reticular connective tissue ,medicine ,Cancer research ,medicine.disease ,Multiple myeloma ,medicine.drug - Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.