32 results on '"Enrico Caserta"'
Search Results
2. CD84 is a regulator of the immunosuppressive microenvironment in multiple myeloma
- Author
-
Hadas Lewinsky, Emine G. Gunes, Keren David, Lihi Radomir, Matthias P. Kramer, Bianca Pellegrino, Michal Perpinial, Jing Chen, Ting-fang He, Anthony G. Mansour, Kun-Yu Teng, Supriyo Bhattacharya, Enrico Caserta, Estelle Troadec, Peter Lee, Mingye Feng, Jonathan Keats, Amrita Krishnan, Michael Rosenzweig, Jianhua Yu, Michael A. Caligiuri, Yosef Cohen, Olga Shevetz, Shirly Becker-Herman, Flavia Pichiorri, Steven Rosen, and Idit Shachar
- Subjects
Hematology ,Oncology ,Medicine - Abstract
Multiple myeloma (MM) is characterized by an accumulation of malignant plasma cells (PCs) within the BM. The BM microenvironment supports survival of the malignant cells and is composed of cellular fractions that foster myeloma development and progression by suppression of the immune response. Despite major progress in understanding the biology and pathophysiology of MM, this disease is still incurable and requires aggressive treatment with significant side effects. CD84 is a self-binding immunoreceptor belonging to the signaling lymphocyte activation molecule (SLAM) family. Previously, we showed that CD84 bridges between chronic lymphocytic leukemia cells and their microenvironment, and it regulates T cell function. In the current study, we investigated the role of CD84 in MM. Our results show that MM cells express low levels of CD84. However, these cells secrete the cytokine macrophage migration inhibitory factor (MIF), which induces CD84 expression on cells in their microenvironment. Its activation leads to an elevation of expression of genes regulating differentiation to monocytic/granulocytic–myeloid-derived suppressor cells (M-MDSCs and G-MDSCs, respectively) and upregulation of PD-L1 expression on MDSCs, which together suppress T cell function. Downregulation of CD84 or its blocking reduce MDSC accumulation, resulting in elevated T cell activity and reduced tumor load. Our data suggest that CD84 might serve as a novel therapeutic target in MM.
- Published
- 2021
- Full Text
- View/download PDF
3. Oncolytic herpes simplex virus infects myeloma cells in vitro and in vivo
- Author
-
Vikas Gupta, Balveen Kaur, James F. Sanchez, Luke Russell, Ji Young Yoo, Steven T. Rosen, Alena Cristina Jaime-Ramirez, Enrico Caserta, Ada Dona, Jayeeta Ghose, Amrita Krishnan, Douglas W. Sborov, Emine Gulsen Gunes, Benjamin G. Barwick, Flavia Pichiorri, Mariam Murtadha, Lawrence H. Boise, and Craig C. Hofmeister
- Subjects
0301 basic medicine ,Cancer Research ,bone marrow ,Biology ,CD38 ,HVEM ,NECTIN-1 ,medicine.disease_cause ,lcsh:RC254-282 ,03 medical and health sciences ,0302 clinical medicine ,malignant plasma cells ,medicine ,Pharmacology (medical) ,Tropism ,oncolytic virus ,apoptosis ,oncolytic herpes simplex virus type 1 (oHSV-1) ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Oncolytic virus ,multiple myeloma ,oncolytic virus (OV) therapy ,030104 developmental biology ,Herpes simplex virus ,medicine.anatomical_structure ,Oncology ,Viral replication ,Apoptosis ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,Molecular Medicine ,Original Article ,Bone marrow ,recombinant virus - Abstract
Because most patients with multiple myeloma (MM) develop resistance to current regimens, novel approaches are needed. Genetically modified, replication-competent oncolytic viruses exhibit high tropism for tumor cells regardless of cancer stage and prior treatment. Receptors of oncolytic herpes simplex virus 1 (oHSV-1), NECTIN-1, and HVEM are expressed on MM cells, prompting us to investigate the use of oHSV-1 against MM. Using oHSV-1-expressing GFP, we found a dose-dependent increase in the GFP+ signal in MM cell lines and primary MM cells. Whereas NECTIN-1 expression is variable among MM cells, we discovered that HVEM is ubiquitously and highly expressed on all samples tested. Expression of HVEM was consistently higher on CD138+/CD38+ plasma cells than in non-plasma cells. HVEM blocking demonstrated the requirement of this receptor for infection. However, we observed that, although oHSV-1 could efficiently infect and kill all MM cell lines tested, no viral replication occurred. Instead, we identified that oHSV-1 induced MM cell apoptosis via caspase-3 cleavage. We further noted that oHSV-1 yielded a significant decrease in tumor volume in two mouse xenograft models. Therefore, oHSV-1 warrants exploration as a novel potentially effective treatment option in MM, and HVEM should be investigated as a possible therapeutic target., Graphical Abstract, Receptors of oncolytic herpes simplex virus (oHSV-1), NECTIN-1, and HVEM are expressed on multiple myeloma (MM) cells. Whereas NECTIN-1 expression is variable among MM cells, HVEM is ubiquitously and highly expressed on all samples tested. However, no viral replication occurred. Instead, oHSV-1 induced MM cell apoptosis via caspase-3 cleavage.
- Published
- 2021
4. A Mathematical Modeling Approach for Targeted Radionuclide and Chimeric Antigen Receptor-T Cell Combination Therapy
- Author
-
John E. Shively, Megan Minnix, Alexander B Brummer, Amrita Krishnan, Xiuli Wang, Russell C. Rockne, Jeffrey Y.C. Wong, Flavia Pichiorri, Vikram Adhikarla, Enrico Caserta, and Dennis Awuah
- Subjects
Cancer Research ,Combination therapy ,medicine.medical_treatment ,T cell ,Cell ,TRT ,actinium-225 ,Article ,combination therapy ,medicine ,Distribution (pharmacology) ,RC254-282 ,Multiple myeloma ,applied_mathematics ,business.industry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Daratumumab ,Immunotherapy ,daratumumab ,medicine.disease ,targeted radionuclide therapy ,Chimeric antigen receptor ,CAR-T ,multiple myeloma ,medicine.anatomical_structure ,Oncology ,Cancer research ,immunotherapy ,CS1 ,alpha particle therapy ,business ,mathematical model - Abstract
Simple Summary Targeted radionuclide therapy (TRT) and immunotherapy, an example being chimeric antigen receptor T cells (CAR-Ts), represent two potent means of eradicating systemic cancers. Although each one as a monotherapy might have a limited effect, the potency can be increased with a combination of the two therapies. The complications involved in the dosing and scheduling of these therapies make the mathematical modeling of these therapies a suitable solution for designing combination treatment approaches. Here, we investigate a mathematical model for TRT and CAR-T cell combination therapies. Through an analysis of the mathematical model, we find that the tumor proliferation rate is the most important factor affecting the scheduling of TRT and CAR-T cell treatments with faster proliferating tumors requiring a shorter interval between the two therapies. Abstract Targeted radionuclide therapy (TRT) has recently seen a surge in popularity with the use of radionuclides conjugated to small molecules and antibodies. Similarly, immunotherapy also has shown promising results, an example being chimeric antigen receptor T cell (CAR-T) therapy in hematologic malignancies. Moreover, TRT and CAR-T therapies possess unique features that require special consideration when determining how to dose as well as the timing and sequence of combination treatments including the distribution of the TRT dose in the body, the decay rate of the radionuclide, and the proliferation and persistence of the CAR-T cells. These characteristics complicate the additive or synergistic effects of combination therapies and warrant a mathematical treatment that includes these dynamics in relation to the proliferation and clearance rates of the target tumor cells. Here, we combine two previously published mathematical models to explore the effects of dose, timing, and sequencing of TRT and CAR-T cell-based therapies in a multiple myeloma setting. We find that, for a fixed TRT and CAR-T cell dose, the tumor proliferation rate is the most important parameter in determining the best timing of TRT and CAR-T therapies.
- Published
- 2021
5. Leflunomide regulates c-Myc expression in myeloma cells through PIM targeting
- Author
-
Domenico Viola, Hongzhi Li, Xiwei Wu, Flavia Pichiorri, James F. Sanchez, Timothy W. Synold, Steven T. Rosen, Ralf Buettner, Michael Rosenzweig, Austin Christofferson, Amrita Krishnan, Jonathan J Keats, Alex E. Pozhitkov, Nagarajan Vaidehi, Estelle Troadec, Joycelynne Palmer, Jihane Khalife, Enrico Caserta, Guido Marcucci, Corey Morales, and Emine Gulsen Gunes
- Subjects
Drug synergism ,Proto-Oncogene Proteins c-myc ,Mice ,chemistry.chemical_compound ,Proto-Oncogene Proteins c-pim-1 ,In vivo ,hemic and lymphatic diseases ,Teriflunomide ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Enzyme Inhibitors ,Lenalidomide ,Active metabolite ,Leflunomide ,Drug Synergism ,Hematology ,Stimulus Report ,chemistry ,Pim kinases ,Cancer research ,Drug Therapy, Combination ,Multiple Myeloma ,Combination method ,medicine.drug - Abstract
Key Points Teriflunomide, the active metabolite of leflunomide, downregulates c-Myc expression through inhibition of PIM kinases. Leflunomide together with lenalidomide significantly extended survival in an in vivo MM model.
- Published
- 2019
6. CD84 is a regulator of the immunosuppressive microenvironment in multiple myeloma
- Author
-
Amrita Krishnan, Michael Rosenzweig, Supriyo Bhattacharya, Mingye Feng, Kun Yu Teng, Jonathan J. Keats, Lihi Radomir, Enrico Caserta, Idit Shachar, Hadas Lewinsky, Michal Perpinial, Shirly Becker-Herman, Yosef Cohen, Ting-Fang He, Steven D. Rosen, Olga Shevetz, Flavia Pichiorri, Estelle Troadec, Keren David, Michael A. Caligiuri, Peter P. Lee, Jing Chen, Matthias P. Kramer, Anthony Mansour, Jianhua Yu, Emine Gulsen Gunes, and Bianca Pellegrino
- Subjects
0301 basic medicine ,medicine.medical_specialty ,T-Lymphocytes ,Chronic lymphocytic leukemia ,medicine.medical_treatment ,T cell ,Cancer immunotherapy ,Lymphocyte Activation ,B7-H1 Antigen ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Downregulation and upregulation ,Signaling Lymphocytic Activation Molecule Family ,Cell Line, Tumor ,Internal medicine ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Macrophage Migration-Inhibitory Factors ,Cancer ,Hematology ,Chemistry ,Myeloid-Derived Suppressor Cells ,Cellular immune response ,General Medicine ,medicine.disease ,Leukemia, Lymphocytic, Chronic, B-Cell ,Intramolecular Oxidoreductases ,030104 developmental biology ,Cytokine ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Medicine ,Macrophage migration inhibitory factor ,Immunotherapy ,Multiple Myeloma ,Research Article - Abstract
Multiple myeloma (MM) is characterized by an accumulation of malignant plasma cells (PCs) within the BM. The BM microenvironment supports survival of the malignant cells and is composed of cellular fractions that foster myeloma development and progression by suppression of the immune response. Despite major progress in understanding the biology and pathophysiology of MM, this disease is still incurable and requires aggressive treatment with significant side effects. CD84 is a self-binding immunoreceptor belonging to the signaling lymphocyte activation molecule (SLAM) family. Previously, we showed that CD84 bridges between chronic lymphocytic leukemia cells and their microenvironment, and it regulates T cell function. In the current study, we investigated the role of CD84 in MM. Our results show that MM cells express low levels of CD84. However, these cells secrete the cytokine macrophage migration inhibitory factor (MIF), which induces CD84 expression on cells in their microenvironment. Its activation leads to an elevation of expression of genes regulating differentiation to monocytic/granulocytic-myeloid-derived suppressor cells (M-MDSCs and G-MDSCs, respectively) and upregulation of PD-L1 expression on MDSCs, which together suppress T cell function. Downregulation of CD84 or its blocking reduce MDSC accumulation, resulting in elevated T cell activity and reduced tumor load. Our data suggest that CD84 might serve as a novel therapeutic target in MM.
- Published
- 2021
7. Comparison of CD38-Targeted α- Versus β-Radionuclide Therapy of Disseminated Multiple Myeloma in an Animal Model
- Author
-
Megan Minnix, Enrico Caserta, John E. Shively, Russell C. Rockne, Flavia Pichiorri, Vikram Adhikarla, and Erasmus Poku
- Subjects
0301 basic medicine ,medicine.medical_specialty ,medicine.drug_class ,medicine.medical_treatment ,Alpha (ethology) ,Pharmacology ,Monoclonal antibody ,Basic Science Investigation ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Radiology, Nuclear Medicine and imaging ,Molecular Targeted Therapy ,Multiple myeloma ,Hematology ,business.industry ,Daratumumab ,Antibodies, Monoclonal ,Radioimmunotherapy ,medicine.disease ,ADP-ribosyl Cyclase 1 ,Beta Particles ,Disease Models, Animal ,030104 developmental biology ,030220 oncology & carcinogenesis ,Radionuclide therapy ,Toxicity ,Safety ,business ,Multiple Myeloma - Abstract
Targeted therapies for multiple myeloma (MM) include the anti-CD38 antibody daratumumab, which, in addition to its inherent cytotoxicity, can be radiolabeled with tracers for imaging and with β- and α-emitter radionuclides for radioimmunotherapy. Methods: We have compared the potential therapeutic efficacy of β- versus α-emitter radioimmunotherapy using radiolabeled DOTA-daratumumab in a preclinical model of disseminated multiple myeloma. Multiple dose levels were investigated to find the dose with the highest efficacy and lowest toxicity. Results: In a dose–response study with the β-emitter (177)Lu-DOTA-daratumumab, the lowest tested dose, 1.85 MBq, extended survival from 37 to 47 d but did not delay tumor growth. Doses of 3.7 and 7.4 MBq extended survival to 55 and 58 d, respectively, causing a small equivalent delay in tumor growth, followed by regrowth. The higher dose, 11.1 MBq, eradicated the tumor but had no effect on survival compared with untreated controls, because of whole-body toxicity. In contrast, the α-emitter (225)Ac-DOTA-daratumumab had a dose-dependent effect, in which 0.925, 1.85, and 3.7 kBq increased survival, compared with untreated controls (35 d), to 47, 52, and 73 d, respectively, with a significant delay in tumor growth for all 3 doses. Higher doses of 11.1 and 22.2 kBq resulted in equivalent survival to 82 d but with significant whole-body toxicity. Parallel studies with untargeted (225)Ac-DOTA-trastuzumab conferred no improvement over untreated controls and resulted in whole-body toxicity. Conclusion: We conclude, and mathematic modeling confirms, that maximal biologic doses were achieved by targeted α-therapy and demonstrated (225)Ac to be superior to (177)Lu in delaying tumor growth and decreasing whole-body toxicity.
- Published
- 2020
8. Identifying CD38+ cells in patients with multiple myeloma: first-in-human imaging using copper-64-labeled daratumumab
- Author
-
Van Eric Biglang-awa, Jeffrey Y.C. Wong, Nitya Nathwani, Flavia Pichiorri, Stephen J. Forman, Michael Rosenzweig, Arnab Chowdhury, Dave Yamauchi, Chatchada Karanes, Guido Marcucci, Russell Rockne, Paul J. Yazaki, Amrita Krishnan, Joycelynne M. Palmer, Anna M. Wu, Nicole Bowles, Enrico Caserta, David Colcher, Erasmus Poku, Vikram Adhikarla, Maria Parayno, Ammar Chaudhry, John E. Shively, Jennifer Simpson, James F. Sanchez, and Firoozeh Sahebi
- Subjects
medicine.diagnostic_test ,Immunobiology and Immunotherapy ,business.industry ,Daratumumab ,Antibodies, Monoclonal ,Hematology ,CD38 ,medicine.disease ,ADP-ribosyl Cyclase 1 ,chemistry.chemical_compound ,Text mining ,chemistry ,Copper Radioisotopes ,Positron emission tomography ,In vivo ,Positron Emission Tomography Computed Tomography ,medicine ,DOTA ,Humans ,Copper-64 ,Nuclear medicine ,business ,Multiple Myeloma ,Multiple myeloma - Abstract
18F-Fluorodeoxyglucose (FDG) positron emission tomography/computed tomography (PET/CT) is one of the most widely used imaging techniques to detect multiple myeloma (MM). Intracellular FDG uptake depicts in vivo metabolic activity, which can be seen in both malignant and nonmalignant cells, resulting in limited sensitivity and specificity. Our group showed preclinically that tracing MM dissemination using a CD38-directed human antibody, daratumumab, that is radioconjugated with 64Cu via the chelator DOTA (64Cu-daratumumab), led to improved sensitivity and specificity over that of FDG. Here, we report the results of a phase 1 trial designed to (1) assess the safety and feasibility of 64Cu-daratumumab PET/CT and (2) preliminarily evaluate and characterize the ability of 64Cu-daratumumab to accurately detect or exclude MM lesions. A total of 12 daratumumab-naive patients were imaged. Prior to the injection of 15 mCi/5 mg of 64Cu-daratumumab, patients were treated with 0 (n = 3), 10 (n = 3), 45 (n = 3), or 95 mg (n = 3) of unlabeled daratumumab to assess its effect on image quality. No significant adverse events were observed from either unlabeled daratumumab or 64Cu-daratumumab. Of the dose levels tested, 45 mg unlabeled daratumumab was the most optimal in terms of removing background signal without saturating target sites. 64Cu-daratumumab PET/CT provided safe whole-body imaging of MM. A trial comparing the sensitivity and specificity of 64Cu-daratumumab PET/CT with that of FDG PET/CT is planned. This trial was registered at www.clinicaltrials.gov as #NCT03311828.
- Published
- 2020
9. Capturing variations in nuclear phenotypes
- Author
-
Gustavo Leone, Kun Huang, Thierry Pécot, Enrico Caserta, Shantanu Singh, Jens Rittscher, Sundaresan Raman, Raghu Machiraju, Department of Computer Science and Engineering [Columbus] (CSE), Ohio State University [Columbus] (OSU), Birla Institute of Technology and Science (BITS Pilani), Broad Institute of MIT and Harvard (BROAD INSTITUTE), Harvard Medical School [Boston] (HMS)-Massachusetts Institute of Technology (MIT)-Massachusetts General Hospital [Boston], Space-timE RePresentation, Imaging and cellular dynamics of molecular COmplexes (SERPICO), Inria Rennes – Bretagne Atlantique, Institut National de Recherche en Informatique et en Automatique (Inria)-Institut National de Recherche en Informatique et en Automatique (Inria), Beckman Research Institute [Duarte, CA], Indiana University School of Medicine, Indiana University System, Institute of Biomedical Engineering [Oxford] (IBME), University of Oxford [Oxford], Hollings Cancer Center [Charleston], Medical University of South Carolina [Charleston] (MUSC), and University of Oxford
- Subjects
General Computer Science ,[SDV.IB.IMA]Life Sciences [q-bio]/Bioengineering/Imaging ,Context (language use) ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,02 engineering and technology ,Computational biology ,Biology ,01 natural sciences ,010305 fluids & plasmas ,Theoretical Computer Science ,0103 physical sciences ,Genotype ,0202 electrical engineering, electronic engineering, information engineering ,medicine ,Gene ,Tumor microenvironment ,[INFO.INFO-CV]Computer Science [cs]/Computer Vision and Pattern Recognition [cs.CV] ,[SDV.BIBS]Life Sciences [q-bio]/Quantitative Methods [q-bio.QM] ,Phenotype ,Chromatin ,medicine.anatomical_structure ,[INFO.INFO-TI]Computer Science [cs]/Image Processing [eess.IV] ,Modeling and Simulation ,020201 artificial intelligence & image processing ,Nucleus ,Biological variability - Abstract
Relating genotypes with phenotypes is important to understand diseases like cancer, but extremely challenging, given the underlying biological variability and levels of phenotypes. 3D quantitative tools are increasingly used to provide robust inferences pertaining to variations across collections of cells. We especially focus on the changes wrought to the nucleus of specific genotypes. Fibroblasts in the tumor microenvironment of mammary epithelial tissue serve as our model system and provide the context, although our methods are applicable to a broader range of biological systems. Using an image based approach, we analyze in 3D and compare phenotypes at nuclear level using estimates of texture, morphology and spatial context based on confocal images. Our data demonstrates that deletion of TP53 in stromal fibroblasts results in reorganization of chromatin content across the nucleus, especially the nuclear periphery, while simultaneously reducing nuclear size and making it more spindly. No such shape change was observed for PTEN-deleted genotype, although there were some differences in distribution of chromatin and an increase in the local nuclear density. The relative changes in phenotypes are in line with the larger role that the TP53 plays in tumor initiation and progression.These findings play an important role in uncovering the relationships of those genes with the subcellular phenotypes, as well as formulating new hypotheses, especially pertaining to the relative impact of genes in specific pathways. More importantly, they demonstrate the efficacy of methodology of analyzing a large number of cellular phenotypes.
- Published
- 2020
10. Daratumumab induces mechanisms of immune activation through CD38+ NK cell targeting
- Author
-
Marianna Martella, Ada Dona, Douglas W. Sborov, Steven T. Rosen, John E. Shively, Lucy Ghoda, Guido Marcucci, Chatchada Karanes, Paul J. Yazaki, James F. Sanchez, Stephen J. Forman, Domenico Viola, Michael Rosenzweig, Jonathan J Keats, Flavia Pichiorri, Myo Htut, Amrita Krishnan, Emine Gulsen Gunes, Arnab Chowdhury, Rodney R. Miles, Xiuli Wang, Tinisha McDonald, Francesca Besi, Jihane Khalife, Enrico Caserta, and Estelle Troadec
- Subjects
CD86 ,0303 health sciences ,education.field_of_study ,Chemistry ,Monocyte ,T cell ,Population ,Priming (immunology) ,CD38 ,Protein degradation ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Immune system ,medicine ,Cancer research ,education ,030304 developmental biology ,030215 immunology - Abstract
Daratumumab (Dara), a multiple myeloma (MM) therapy, is an antibody against the surface receptor CD38, which is expressed not only on plasma cells but also on NK cells and monocytes. Correlative data have highlighted the immune-modulatory role of Dara, despite the paradoxical observation that Dara regimens decrease the frequency of total NK cells. Here we show that, despite this reduction, NK cells play a pivotal role in Dara anti-MM activity. CD38 on NK cells is essential for Dara-induced immune modulation, and its expression is restricted to NK cells with effector function. We also show that Dara induces rapid CD38 protein degradation associated with NK cell activation, leaving an activated CD38-negative NK cell population. CD38+ NK cell targeting by Dara also promotes monocyte activation, inducing an increase in T cell costimulatory molecules (CD86/80) and enhancing anti-MM phagocytosis activity ex-vivo and in vivo. In support of Dara’s immunomodulating role, we show that MM patients that discontinued Dara therapy because of progression maintain targetable unmutated surface CD38 expression on their MM cells, but retain effector cells with impaired cellular immune function. In summary, we report that CD38+ NK cells may be an unexplored therapeutic target for priming the immune system of MM patients.
- Published
- 2019
- Full Text
- View/download PDF
11. MiR-16 regulates crosstalk in NF-κB tolerogenic inflammatory signaling between myeloma cells and bone marrow macrophages
- Author
-
Amrita Krishnan, Marta Chesi, Enrico Caserta, Estelle Troadec, Domenico Viola, Emine Gulsen Gunes, Guido Marcucci, Abhay R. Satoskar, Alex E. Pozhitkov, Jayeeta Ghose, Alberto Rocci, Jihane Khalife, Cesar Terrazas, Steven D. Rosen, James F. Sanchez, Ada Dona, Marianna Martella, P. Leif Bergsagel, Craig C. Hofmeister, Jonathan J. Keats, and Flavia Pichiorri
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Down-Regulation ,Bone Marrow Cells ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Internal medicine ,Cell Line, Tumor ,medicine ,Extracellular ,Tumor Microenvironment ,Animals ,Humans ,Mice, Knockout ,Hematology ,Chemistry ,Cell growth ,Macrophages ,NF-kappa B ,NF-κB ,General Medicine ,Crosstalk (biology) ,MicroRNAs ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Proteasome inhibitor ,Cancer research ,Bone marrow ,Multiple Myeloma ,Intracellular ,medicine.drug ,Research Article ,Signal Transduction - Abstract
High levels of circulating miR-16 in the serum of multiple myeloma (MM) patients are independently associated with longer survival. Although the tumor suppressor function of intracellular miR-16 in MM plasma cells (PCs) has been elucidated, its extracellular role in maintaining a nonsupportive cancer microenvironment has not been fully explored. Here, we show that miR-16 is abundantly released by MM cells through extracellular vesicles (EVs) and that differences in its intracellular expression as associated with chromosome 13 deletion (Del13) are correlated to extracellular miR-16 levels. We also demonstrate that EVs isolated from MM patients and from the conditioned media of MM-PCs carrying Del13 more strongly differentiate circulating monocytes to M2-tumor supportive macrophages (TAMs), compared with MM-PCs without this chromosomal aberration. Mechanistically, our data show that miR-16 directly targets the IKKα/β complex of the NF-κB canonical pathway, which is critical not only in supporting MM cell growth, but also in polarizing macrophages toward an M2 phenotype. By using a miR-15a-16-1-KO mouse model, we found that loss of the miR-16 cluster supports polarization to M2 macrophages. Finally, we demonstrate the therapeutic benefit of miR-16 overexpression in potentiating the anti-MM activity by a proteasome inhibitor in the presence of MM-resident bone marrow TAM.
- Published
- 2019
12. Caserta E, Chea J, Minnix M, et al. Copper 64–labeled daratumumab as a PET/CT imaging tracer for multiple myeloma. Blood. 2018;131(7):741-745
- Author
-
John E. Shively, Susanta K. Hui, Paul J. Yazaki, Junie Chea, Domenico Viola, Jihane Khalife, Amrita Krishnan, Steven Vonderfecht, Desiree Crow, Joycelynne Palmer, Guido Marcucci, Young L. Kim, James F. Sanchez, Jonathan J Keats, Flavia Pichiorri, Megan Minnix, Erasmus Poku, David Colcher, Nadia Carlesso, Steven D. Rosen, Ralf Buettner, and Enrico Caserta
- Subjects
Immunology ,Pet ct imaging ,Biochemistry ,Mice ,TRACER ,Cell Line, Tumor ,Positron Emission Tomography Computed Tomography ,medicine ,Animals ,Humans ,Radioactive Tracers ,Multiple myeloma ,business.industry ,Daratumumab ,Antibodies, Monoclonal ,Cell Biology ,Hematology ,medicine.disease ,Copper Radioisotopes ,Cell Tracking ,Heterografts ,Copper-64 ,Erratum ,Nuclear medicine ,business ,Multiple Myeloma ,Neoplasm Transplantation ,Half-Life - Abstract
As a growing number of patients with multiple myeloma (MM) respond to upfront therapies while eventually relapsing in a time frame that is often unpredictable, attention has increasingly focused on developing novel diagnostic criteria to also account for disease dissemination. Positron emission tomography/computed tomography (PET/CT) is often used as a noninvasive monitoring strategy to assess cancer cell dissemination, but because the uptake of the currently used radiotracer 18fluorodeoxyglucose (
- Published
- 2018
13. Copper 64–labeled daratumumab as a PET/CT imaging tracer for multiple myeloma
- Author
-
Megan Minnix, Domenico Viola, Erasmus Poku, Flavia Pichiorri, Young L. Kim, Paul J. Yazaki, James F. Sanchez, Junie Chea, Enrico Caserta, Jonathan J Keats, John E. Shively, Susanta K. Hui, Joycelynne Palmer, Ralf Buettner, Jihane Khalife, Guido Marcucci, Nadia Carlesso, Steven D. Rosen, Desiree Crow, David Colcher, Amrita Krishnan, and Steven Vonderfecht
- Subjects
0301 basic medicine ,Immunobiology and Immunotherapy ,Immunology ,Cell ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,immune system diseases ,hemic and lymphatic diseases ,medicine ,DOTA ,neoplasms ,Multiple myeloma ,biology ,medicine.diagnostic_test ,business.industry ,Daratumumab ,Cell Biology ,Hematology ,medicine.disease ,Imaging agent ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Positron emission tomography ,030220 oncology & carcinogenesis ,Cancer cell ,biology.protein ,Antibody ,business ,Nuclear medicine - Abstract
As a growing number of patients with multiple myeloma (MM) respond to upfront therapies while eventually relapsing in a time frame that is often unpredictable, attention has increasingly focused on developing novel diagnostic criteria to also account for disease dissemination. Positron emission tomography/computed tomography (PET/CT) is often used as a noninvasive monitoring strategy to assess cancer cell dissemination, but because the uptake of the currently used radiotracer 18fluorodeoxyglucose (18F-FDG) is a function of the metabolic activity of both malignant and nonmalignant cells, the results frequently lack sufficient specificity. Radiolabeled antibodies targeting MM tissue may detect disease irrespective of cell metabolism. Hence, we conjugated the clinically significant CD38-directed human antibody daratumumab (Darzalex [Dara]) to the DOTA chelator and labeled it with the positron-emitting radionuclide copper 64 (64Cu; 64Cu-DOTA-Dara). Here, we show that 64Cu-DOTA-Dara can efficiently bind CD38 on the surface of MM cells and was mainly detected in the bones associated with tumor in a MM murine model. We also show that PET/CT based on 64Cu-DOTA-Dara displays a higher resolution and specificity to detect MM cell dissemination than does 18F-FDG PET/CT and was even more sensitive than were bioluminescence signals. We therefore have supporting evidence for using 64Cu-DOTA-Dara as a novel imaging agent for MM.
- Published
- 2018
14. CD84: A Potential Novel Therapeutic Target in the Multiple Myeloma Microenvironment
- Author
-
James F. Sanchez, Enrico Caserta, Jonathan J. Keats, Guido Marcucci, Ralf Buettner, Idit Shachar, Hadas Lewinsky, Myo Htut, Flavia Pichiorri, Domenico Viola, Michael Rosenzweig, Emine Gulsen Gunes, and Steven D. Rosen
- Subjects
Cancer Research ,Oncology ,business.industry ,Cancer research ,Medicine ,Hematology ,business ,medicine.disease ,Multiple myeloma - Published
- 2019
15. Reolysin and Histone Deacetylase Inhibition in the Treatment of Head and Neck Squamous Cell Carcinoma
- Author
-
Quintin Pan, John H. Lee, Alena Cristina Jaime-Ramirez, Theodoros N. Teknos, Bhavna Kumar, Enrico Caserta, Flavia Pichiorri, Robert A. Baiocchi, Jianying Zhang, Tae Jin Lee, Pawan Kumar, Jun Ge Yu, Ji Young Yoo, Craig C. Hofmeister, Balveen Kaur, and Matthew O. Old
- Subjects
0301 basic medicine ,Cancer Research ,Combination therapy ,Cell ,oncolytics ,Biology ,reovirus ,lcsh:RC254-282 ,03 medical and health sciences ,0302 clinical medicine ,Reolysin ,medicine ,Pharmacology (medical) ,Vorinostat ,Head and neck cancer ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Head and neck squamous-cell carcinoma ,Virology ,immune impact ,3. Good health ,Oncolytic virus ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Molecular Medicine ,Original Article ,head and neck cancer ,Histone deacetylase ,medicine.drug - Abstract
Oncolytic viruses (OVs) are emerging as powerful anti-cancer agents and are currently being tested for their safety and efficacy in patients. Reovirus (Reolysin), a naturally occurring non-pathogenic, double-stranded RNA virus, has natural oncolytic activity and is being tested in phase I–III clinical trials in a variety of tumor types. With its recent US Food and Drug Administration (FDA) orphan drug designation for several tumor types, Reolysin is a potential therapeutic agent for various cancers, including head and neck squamous cell carcinomas (HNSCCs), which have a 5-year survival of ∼55%. Histone deacetylase inhibitors (HDACis) comprise a structurally diverse class of compounds with targeted anti-cancer effects. The first FDA-approved HDACi, vorinostat (suberoylanilide hydroxamic acid [SAHA]), is currently being tested in patients with head and neck cancer. Recent findings indicate that HDAC inhibition in myeloma cells results in the upregulation of the Reolysin entry receptor, junctional adhesion molecule 1 (JAM-1), facilitating reovirus infection and tumor cell killing both in vitro and in vivo. In this study, we tested the anti-tumor efficacy of HDAC inhibitors AR-42 or SAHA in conjunction with Reolysin in HNSCCs. While HDAC inhibition increased JAM-1 and reovirus entry, the impact of this combination therapy was tested on the development of anti-tumor immune responses.
- Published
- 2017
16. First-in-Human Imaging of Multiple Myeloma Using Copper-64-Labeled Daratumumab: Preliminary Results
- Author
-
Stephen J. Forman, Arnab Chowdhury, John E. Shively, Michael Rosenzweig, Vikram Adhikarla, Joycelynne Palmer, Erasmus Poku, Amrita Krishnan, Firoozeh Sahebi, Maria Parayno, David Colcher, Chatchada Karanes, Van Eric Biglang-awa, James F. Sanchez, Anna M. Wu, Jennifer Simpson, Enrico Caserta, David Yamauchi, Flavia Pichiorri, Nicole Bowles, Paul J. Yazaki, Nitya Nathwani, and Ammar Chaudhry
- Subjects
business.industry ,Immunology ,Whole body imaging ,Daratumumab ,Cell Biology ,Hematology ,First in human ,medicine.disease ,Biochemistry ,medicine ,Medical imaging ,Plasmacytoma ,Copper-64 ,Nuclear medicine ,business ,neoplasms ,Multiple myeloma ,Minimal cognitive impairment - Abstract
Introduction: 18Fluoro-deoxyglucose (FDG) positron emission tomography/computed tomography (PET/CT) is one of the most widely used imaging techniques to detect multiple myeloma (MM). Intracellular FDG uptake depicts in vivo metabolic activity, which can be seen in both malignant and non-malignant cells, resulting in limited sensitivity and specificity. Our group showed preclinically that tracing MM dissemination using a CD38-directed human antibody, daratumumab, that is radioconjugated with copper-64 via the chelator DOTA (64Cu-DOTA-Dara), led to improved sensitivity and specificity over that of FDG (Caserta et al, Blood 2018). Herein, we report the results of a Phase 1 trial (NCT#03311828) designed to 1) assess the safety and feasibility of 64Cu-DOTA-Dara PET/CT and 2) to evaluate and characterize the ability of 64Cu-DOTA-Dara to accurately detect or exclude MM lesions compared with FDG PET/CT. Methods: Patients with biopsy-proven MM and/or a plasmacytoma received an FDG PET/CT scan within 60 days of enrollment. On Day 0, patients were infused with unlabeled ("cold") Dara at one of four dose levels (0 mg, 10 mg, 45 mg, 95 mg) to optimize biodistribution of radioconjugated 64Cu-DOTA-Dara, especially in the liver and the spleen. Within 6 hours of unlabeled Dara administration, patients received 64Cu-DOTA-Dara at a dose of 13.63-16.68 mCi (~5 mg). Whole-body PET scans were obtained at 24 hours and at 48 hours (the latter scan encompassing known tumors). 64Cu-DOTA-Dara standardized uptake values (SUV) were evaluated in MM lesions and normal organs, which were then compared with values from standard FDG PET/CT. Biopsies were performed on accessible discordant lesions. Results: A total of 10 Dara-naïve patients were imaged. Patients were treated with 0 (n=3), 10 (n=3), 45 (n=3), or 95 mg (n=1) of unlabeled Dara. Four patients had newly diagnosed disease, one had biochemical relapse, one had a recurrent plasmacytoma by MRI, and four had possible recurrence by standard PET/CT. No significant adverse events were observed from either cold or 64Cu-DOTA-Dara. With the exception of the one patient with a recurrent plasmacytoma, radiolabeled antibody was eliminated from systemic circulation in subjects analyzed at the first three dose levels within 30 min post injection. In the patient with a recurrent plasmacytoma, the radiolabeled antibody was elevated in the blood for over two days. One newly diagnosed patient had extensive disease by FDG PET and had a biopsy-proven target lesion in the sternum, which had an SUVmax of 14.7 on 64Cu-DOTA-Dara PET/CT vs. 3.3 onFDG PET/CT. A second biopsy from the same patient was taken from a discordant iliac crest lesion (positive for 64Cu-DOTA-Dara but negative for FDG PET/CT) that showed 20-30% MM cell infiltration. In another patient, an iliac crest lesion was 64Cu-DOTA-Dara positive and FDG-negative; biopsy revealed 6% plasmacytosis in the bone. A third patient had an FDG PET/CT positive pleural lesion with an SUVmax of 8.98 and negative on 64Cu-DOTA-Dara (Figure 1A). The lesion did not show recurrence upon biopsy. Furthermore, 64Cu-DOTA-Dara PET/CT yielded superior imaging of bone lesions in the calvarium (Figure 1B). Escalating doses of unlabeled Dara decreased liver and spleen uptake by 64Cu-DOTA-Dara. Conclusions: In this ongoing study, 64Cu-DOTA-Dara PET/CT imaging is to date safe and provides whole body imaging of MM. Further dose escalation of cold Dara (145 mg, 195 mg) is planned to optimize background interference. This modality has the potential to improve sensitivity and specificity over FDG PET/CT scanning in early-stage MM as well as in recurrent disease. Disclosures Krishnan: Celgene, Janssen, Sanofi, BMS: Consultancy; Sutro BioPharma, zPredicta: Consultancy; Amgen, Takeda: Speakers Bureau; Celgene, Z Predicta: Other: Stock Ownership; Takeda: Research Funding. Palmer:Gilead Sciences: Consultancy. Rosenzweig:Celgene, Takeda: Speakers Bureau. Wu:ImaginAb, Inc.: Consultancy, Other: Board Member.
- Published
- 2019
17. Proteasome Inhibitors Impair the Innate Antiviral Immune Response and Potentiate Pelareorep-Based Viral Therapy in Multiple Myeloma
- Author
-
Amrita Krishnan, Matthew C. Coffey, James F. Sanchez, Ada Dona, Douglas W. Sborov, Flavia Pichiorri, Francesca Besi, Craig C. Hofmeister, Jonathan J Keats, Guido Marcucci, Gerard J. Nuovo, Enrico Caserta, and Domenico Viola
- Subjects
Bortezomib ,business.industry ,medicine.medical_treatment ,Immunology ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Carfilzomib ,chemistry.chemical_compound ,medicine.anatomical_structure ,Immune system ,Cytokine ,Proteasome ,chemistry ,Cell culture ,medicine ,Cancer research ,Bone marrow ,business ,Multiple myeloma ,medicine.drug - Abstract
INTRODUCTION: Pelareorep is the infusible form of human reovirus (RV). Our single-agent phase 1 RV trial in relapsed multiple myeloma (MM) showed that pelareorep treatment selectively infected MM cells, as viral RNA was found in myeloma cells and not the bone marrow (BM) stroma. However, we did not observe apoptosis. Our ongoing phase 1 trial, which combines the proteasome inhibitor carfilzomib with RV, has demonstrated RV infection, apoptosis, and clinical responses. We investigated the molecular mechanisms behind the role of a PI (carfilzomib) in this setting. RESULTS: In all MM cell lines we tested (n=4), independently from their sensitivity to RV infection (Stiff et al., Mol Cancer Ther, 2016), viral replication and apoptosis was impaired when MM cells were directly exposed to PIs (carfilzomib and bortezomib). When this experiment was repeated in the setting of the bone marrow cellular fraction or peripheral blood mononuclear cells (PBMCs), it had the opposite effect, as the addition of PI to RV increased RV replication and apoptosis in MM cells. When we washed PBMCs after overnight exposure to RV+PI or to either single agent, then added MM cells, we observed higher infection and apoptosis in cancer cells co-cultured with RV+PI compared to levels from PBMCs treated with each of the single agents, suggesting that PIs increase the ability of PBMCs to serve as a reservoir for infectious reovirus. Monocytes (CD14+) can engage in phagocytosis of reovirus (Berkeley et al., Cancer Immunol Res, 2018), and accordingly we found that RV genome and capsid protein production were detected in CD14+ cells, but not in CD14-depleted PBMCs, and increased upon PI treatment compared to that in RV-treated CD14+ cells. Given that the NF-κB complex is a key proinflammatory signaling pathway associated with the early innate-antiviral immune response, and because PIs can block the degradation of the NF-κB inhibitor IκBα upon phosphorylation, we investigated the effect of the specific IκBα inhibitor Bay-11 in RV viral replication. Our data show that either PI or Bay-11 can inhibit RV-induced IκBα phosphorylation and its subsequent degradation upon RV infection in CD14+ cells, an effect associated with higher capsid formation in RV-treated CD14+ cells in combination with PI or Bay-11, compared to levels from RV alone. Cytokine profiling in PBMCs and CD14+ cells treated with RV in combination with either PI or Bay-11 showed a significant decrease in IFN-α and IFN-β (IFNs) levels and a concomitant increase in RV replication, in contrast to levels from RV alone (p We then decided to investigate whether CD14 depletion could affect RV delivery to the cancer cells invivo. Upon intra-femoral injection of 5TGM1 MM cells into syngeneic C57BL/KaLwRij mice, the mice in which the monocytes were depleted by clodronate-liposome treatment before intravenous RV injection showed lower capsid protein formation in the BM MM cells compared to that in mice where the monocytic population was intact. Because monocytes respond to infection by dividing into macrophages to eliminate pathogens, we wanted to investigate whether PI could impair this effect. Intriguingly, although higher levels of viral genome were detected in PI+RV-treated CD14+ cells compared to RV-treated cells (p CONCLUSIONS: Here we report for the first time that PIs enhance pelareorep entry, infection, and killing of myeloma cells through its effect on the CD14+ fraction. Reovirus infection and replication within CD14+ cells are augmented by PI-induced NF-κB inhibition of the early innate pro-inflammatory immune response. We also report for the first time that carfilzomib induces direct T cell activation and potentiates T cell killing activity against RV-infected MM cells. Disclosures Krishnan: Takeda: Research Funding; Celgene, Z Predicta: Other: Stock Ownership; Amgen, Takeda: Speakers Bureau; Sutro BioPharma, zPredicta: Consultancy; Celgene, Janssen, Sanofi, BMS: Consultancy. Sborov:Celgene: Honoraria; Janssen: Consultancy. Hofmeister:Nektar: Honoraria, Membership on an entity's Board of Directors or advisory committees; Karyopharm: Membership on an entity's Board of Directors or advisory committees; Imbrium: Membership on an entity's Board of Directors or advisory committees; Celgene: Honoraria; Janssen: Membership on an entity's Board of Directors or advisory committees; Oncopeptides: Membership on an entity's Board of Directors or advisory committees.
- Published
- 2019
18. Histone Deacetylase Inhibitors Enhance the Therapeutic Potential of Reovirus in Multiple Myeloma
- Author
-
Douglas W. Sborov, Sarah Schlotter, Alena Cristina Jaime-Ramirez, Joseph Badway, Emily Smith, John C. Byrd, Craig C. Hofmeister, Alessandro Canella, Enrico Caserta, Gerard J. Nuovo, Xiaokui Mo, Matthew O. Old, Don M. Benson, Andrew Stiff, Flavia Pichiorri, Pearlly S. Yan, Robert A. Baiocchi, and Balveen Kaur
- Subjects
0301 basic medicine ,Male ,Cancer Research ,Virus genetics ,Genetic Vectors ,Gene Expression ,Antineoplastic Agents ,Receptors, Cell Surface ,Biology ,Article ,Epigenesis, Genetic ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Cell Line, Tumor ,Reolysin ,medicine ,Animals ,Humans ,Multiple myeloma ,Oncolytic Virotherapy ,Cancer ,medicine.disease ,Combined Modality Therapy ,Xenograft Model Antitumor Assays ,Oncolytic virus ,Tumor Burden ,Histone Deacetylase Inhibitors ,Disease Models, Animal ,Oncolytic Viruses ,030104 developmental biology ,Histone ,Oncology ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,biology.protein ,Receptors, Virus ,Histone deacetylase ,Multiple Myeloma ,Cell Adhesion Molecules - Abstract
Multiple myeloma remains incurable and the majority of patients die within 5 years of diagnosis. Reolysin, the infusible form of human reovirus (RV), is a novel viral oncolytic therapy associated with antitumor activity likely resulting from direct oncolysis and a virus-mediated antitumor immune response. Results from our phase I clinical trial investigating single agent Reolysin in patients with relapsed multiple myeloma confirmed tolerability, but no objective responses were evident, likely because the virus selectively entered the multiple myeloma cells but did not actively replicate. To date, the precise mechanisms underlying the RV infectious life cycle and its ability to induce oncolysis in patients with multiple myeloma remain unknown. Here, we report that junctional adhesion molecule 1 (JAM-1), the cellular receptor for RV, is epigenetically regulated in multiple myeloma cells. Treatment of multiple myeloma cells with clinically relevant histone deacetylase inhibitors (HDACi) results in increased JAM-1 expression as well as increased histone acetylation and RNA polymerase II recruitment to its promoter. Furthermore, our data indicate that the combination of Reolysin with HDACi, potentiates RV killing activity of multiple myeloma cells in vitro and in vivo. This study provides the molecular basis to use these agents as therapeutic tools to increase the efficacy of RV therapy in multiple myeloma. Mol Cancer Ther; 15(5); 830–41. ©2016 AACR.
- Published
- 2016
19. CD84: A Potential Novel Therapeutic Target in Multiple Myeloma
- Author
-
Emine Gulsen Gunes, Domenico Viola, Flavia Pichiorri, Jonathan J Keats, Amrita Krishnan, Idit Shachar, Ralf Buettner, Steven T. Rosen, Hadas Lewinsky, Enrico Caserta, Myo Htut, James F. Sanchez, Michael Rosenzweig, and Guido Marcucci
- Subjects
Cell type ,education.field_of_study ,Myeloid ,Chemistry ,T cell ,Immunology ,Population ,Cell Biology ,Hematology ,Biochemistry ,Molecular biology ,medicine.anatomical_structure ,Cell culture ,Myeloid-derived Suppressor Cell ,medicine ,Bone marrow ,Receptor ,education - Abstract
Introduction: Multiple Myeloma (MM) cell survival strictly depends on the interaction with multiple cell types in the bone marrow (BM), collectively referred to as the MM microenvironment (MM-ME). CD84 (SLAMF5) belongs to the signaling lymphocyte activation molecule family of immunoreceptors; previous data from our group have shown that CD84 mediates malignant B cells and their ME (Marom et al. 2017); however, its role within the MM-ME has not yet been investigated. Results: Using the MMRF CoMMpass IA9 dataset, we found that CD84 mRNA expression is low or absent in CD138+MM cells isolated from 660 newly diagnosed patients. In agreement with these data, flow analysis showed an absence of CD84 expression in all MM cell lines tested (n=9) and minimal surface expression (5.5-13%) in primary cells (n=3). However, a significantly higher expression of CD84 was detected on the surface of BM and peripheral blood (PB) monocytic fractions (CD14+) (76-97%) compared to that of matched CD14 negative fractions (2-9%) obtained from 16 different MM patients (p Next, to understand whether the expansion of Mo-MDSC and associated CD84 up-regulation is directly dependent on the MM cells, we investigated Mo-MDSC expansion and CD84 expression in two different MM mouse models. Specifically, 5TGM1 murine MM cells were IV injected into syngeneic immune-competent KaLwRijHsd mice (n=7), and human MM.1S cells were IV injected into immune-deficient NSG mice (n=10). Our data show that MM progression leads to significant Mo-MDSC expansion (p Since T cell checkpoint inhibitors PD-1/PD-L1 are tumor immune escape receptors that play a pivotal role in supporting an immunosuppressive MM-ME, we also investigated the expression of PD-L1 on the CD14+ fraction in MM patients. BM-CD14+ cells (n=3) were compared with matched CD14 negative cells. We found that the CD14+ fraction had a higher PD-L1 expression in the BM of MM patients compared to the CD14 negative fraction (60-96% versus 3-8%). Hence, we investigated whether CD84-mediated cell-cell contact may increase the level of PD-L1 expression on Mo-MDSCs and whether MM cells can regulate CD84 and PD-L1 expression on CD14+ cells. Co-culture assays were performed using several human MM cell lines (MM1S, U266 and KMS11) with PB CD14+ cells derived from healthy donors (n=3). Our data show a 1.9-fold increase from the basal level of CD84 (27±16% to 50±9.4%) and a 2.9-fold increase from the basal level of PD-L1 (18±8.5% to 52.3±5.7%). Our recently generated mouse anti-human CD84 blocking antibody (Marom et al. 2017) was used to determine whether CD84 direct targeting can affect MM-induced CD84 and PD-L1 expression on the surface of CD14+cells. Co-culture experiments show that targeting CD84 lowered CD84 (1.6-fold decrease) and PD-L1 expression (1.8-fold decrease) on a CD14+ population co-cultured with MM cells. Moreover, when T cells were included in the co-culture experiments, we observed reduced expression of exhaustion markers PD-1 (1.1-fold decrease) and CTLA4 (1.4-fold decrease) in the presence of the CD84 blocking antibody. Conclusion: We show here that CD84 is expressed on Mo-MDSCs in MM and that the expansion of this myeloid population is strictly associated with MM progression. Disruption of CD84 contacts with a blocking antibody decreases the expression of PD-L1 on CD14+ cells and exhaustion markers on T cells. Thus, our results reveal a novel role for CD84 in regulating PD-L1 in the MM-ME and provides the scientific rationale to investigate whether targeting CD84 expression on Mo-MDSCs can restore T cell functions. To further confirm the role of CD84 regulating T cell activity and Mo-MDSC, in vivo mouse studies are ongoing and results will be presented at the meeting. Disclosures Rosenzweig: Celgene: Speakers Bureau. Krishnan:Sutro: Speakers Bureau; Onyx: Speakers Bureau; Janssen: Consultancy, Speakers Bureau; Celgene: Consultancy, Equity Ownership, Speakers Bureau; Takeda: Speakers Bureau.
- Published
- 2018
20. Daratumumab induces CD38 internalization and impairs myeloma cell adhesion
- Author
-
Domenico Viola, Jonathan J Keats, Cesar Terrazas, Michael Rosenzweig, Guido Marcucci, Enrico Caserta, Flavia Pichiorri, Estelle Troadec, Tinisha McDonald, Balveen Kaur, Jayeeta Ghose, Steven D. Rosen, Emine Gulsen Gunes, James F. Sanchez, Amrita Krishnan, Abhay R. Satoskar, Craig C. Hofmeister, and Jihane Khalife
- Subjects
lcsh:Immunologic diseases. Allergy ,0301 basic medicine ,media_common.quotation_subject ,Immunology ,CD38 ,lcsh:RC254-282 ,plasma cells ,cd38 ,03 medical and health sciences ,In vivo ,medicine ,Immunology and Allergy ,Internalization ,Cell adhesion ,Original Research ,media_common ,Bortezomib ,Chemistry ,bortezomib ,loss of adhesion ,Daratumumab ,daratumumab ,sensitivity ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,multiple myeloma ,internalization ,030104 developmental biology ,Oncology ,Proteasome ,Monoclonal ,Cancer research ,lcsh:RC581-607 ,bone marrow stromal cells ,medicine.drug - Abstract
Daratumumab (Dara), a human immunoglobulin G1 kappa (IgG1κ) monoclonal anti-CD38 antibody, has been approved by the U.S. Food and Drug Administration for the treatment of relapsed multiple myeloma (MM) as a single agent as well as in combination with immunomodulatory drugs (IMiDs) and proteasome inhibitors (PI). Although the scientific rationale behind the use of Dara in combination with IMiDs has been extensively explored, the molecular mechanisms underlying Dara-PI regimens have not yet been investigated. Here, we demonstrate that CD38 on the surface of MM cells is rapidly internalized after Dara treatment; we also show that Dara treatment impairs MM cell adhesion, an effect that can be rescued by using the endocytosis inhibitor Dynasore. Finally, we show that Dara potentiates bortezomib (BTZ) killing of MM cells in vitro and in vivo, independent of its function as an immune activator. In conclusion, our data show that Dara impairs MM cell adhesion, which results in an increased sensitivity of MM to proteasome inhibition.
- Published
- 2018
21. Allele-specific tumor spectrum in Pten knockin mice
- Author
-
Shan Naidu, Gustavo Leone, Wolfgang Sadee, Hui-Zi Chen, Paul C. Stromberg, Hui Wang, Matt Karikomi, Maysoon Rawahneh, Thomas J. Rosol, Charis Eng, Danxin Wang, Ravi Rajmohan, Julie A. Stephens, Michael Weinstein, Michael C. Ostrowski, Enrico Caserta, Krista M. D. La Perle, Julie Moffitt, and Soledad Fernandez
- Subjects
Tumor suppressor gene ,DNA Mutational Analysis ,Molecular Sequence Data ,Embryonic Development ,Biology ,Mice ,Germline mutation ,Neoplasms ,medicine ,Animals ,Point Mutation ,Tensin ,Missense mutation ,PTEN ,Genetic Predisposition to Disease ,Gene Knock-In Techniques ,Gene Silencing ,Alleles ,Cell Proliferation ,Genetics ,Multidisciplinary ,Base Sequence ,Protein Stability ,Tumor Suppressor Proteins ,Point mutation ,PTEN Phosphohydrolase ,Cowden syndrome ,Biological Sciences ,medicine.disease ,Organ Specificity ,Lipid phosphatase activity ,Gene Targeting ,Disease Progression ,Embryo Loss ,biology.protein ,Mutant Proteins ,Precancerous Conditions ,Proto-Oncogene Proteins c-akt - Abstract
Germline mutations in the tumor suppressor gene PTEN (phosphatase and tensin homology deleted on chromosome 10) cause Cowden and Bannayan–Riley–Ruvalcaba (BRR) syndromes, two dominantly inherited disorders characterized by mental retardation, multiple hamartomas, and variable cancer risk. Here, we modeled three sentinel mutant alleles of PTEN identified in patients with Cowden syndrome and show that the nonsense Pten ∆4–5 and missense Pten C124R and Pten G129E alleles lacking lipid phosphatase activity cause similar developmental abnormalities but distinct tumor spectra with varying severity and age of onset. Allele-specific differences may be accounted for by loss of function for Pten ∆4–5 , hypomorphic function for Pten C124R , and gain of function for Pten G129E . These data demonstrate that the variable tumor phenotypes observed in patients with Cowden and BRR syndromes can be attributed to specific mutations in PTEN that alter protein function through distinct mechanisms.
- Published
- 2010
22. Ribosomal Interaction of Bacillus stearothermophilus Translation Initiation Factor IF2: Characterization of the Active Sites
- Author
-
Carlotta Ferrara, Pohl Milón, Jerneja Tomšic, Enrico Caserta, Alessandra Rocchetti, Claudio O. Gualerzi, Anna La Teana, Cynthia L. Pon, and Attilio Fabbretti
- Subjects
Ribosomal Interaction ,Mutant ,GTPase ,Prokaryotic Initiation Factor-2 ,Biology ,medicine.disease_cause ,Ribosome ,Protein Structure, Secondary ,Geobacillus stearothermophilus ,Structural Biology ,Catalytic Domain ,Escherichia coli ,Ribosome Subunits ,medicine ,Initiation factor ,Molecular Biology ,Sequence Deletion ,Genetics ,Mutation ,Genetic Complementation Test ,Dipeptides ,Ribosomal RNA ,Null allele ,Kinetics ,Amino Acid Substitution ,Genes, Bacterial ,Protein Biosynthesis ,Mutant Proteins ,Ribosomes ,Protein Binding - Abstract
InfB-encoded translation initiation factor IF2 contains a non-conserved N-terminal domain and two conserved domains (G and C) constituted by three (G1, G2 and G3) and two (C1 and C2) sub-domains. Here, we show that: (i) Bacillus stearothermophilus IF2 complements in vivo an Escherichia coli infB null mutation and (ii) the N-domain of B. stearothermophilus IF2, like that of E. coli IF2, provides a strong yet dispensable interaction with 30 S and 50 S subunits in spite of the lack of any size, sequence or structural homology between the N-domains of the two factors. Furthermore, the nature of the B. stearothermophilus IF2 sites involved in establishing the functional interactions with the ribosome was investigated by generating deletion, random and site-directed mutations within sub-domains G2 or G3 of a molecule carrying an H301Y substitution in switch II of the G2 module, which impairs the ribosome-dependent GTPase activity of IF2. By selecting suppressors of the dominant-lethal phenotype caused by the H301Y substitution, three independent mutants impaired in ribosome binding were identified; namely, S387P (in G2) and G420E and E424K (in G3). The functional properties of these mutants and those of the deletion mutants are compatible with the premise that IF2 interacts with 30 S and 50 S subunits via G3 and G2 modules, respectively. However, beyond this generalization, because the mutation in G2 resulted in a functional alteration of G3 and vice versa, our results indicate the existence of extensive "cross-talking" between these two modules, highlighting a harmonic conformational cooperation between G2 and G3 required for a functional interaction between IF2 and the two ribosomal subunits. It is noteworthy that the E424K mutant, which completely lacks GTPase activity, displays IF2 wild-type capacity in supporting initiation of dipeptide formation.
- Published
- 2010
23. Pten in stromal fibroblasts suppresses mammary epithelial tumours
- Author
-
Julie A. Stephens, Michael C. Ostrowski, Fu Li, Shan Naidu, Hui Wang, Jean-Leon Chong, Metin N. Gurcan, Gustavo Leone, Carmen Z. Cantemir-Stone, Nicholas Creasap, Michael Hallett, Sudarshana M. Sharma, Francois Pepin, Soledad Fernandez, Lisa D. Yee, John C. Thompson, Enrico Caserta, Anand S. Merchant, Thomas J. Rosol, Julie A. Wallace, Michael Weinstein, Anthony J. Trimboli, Morag Park, Michael L. Robinson, Paul C. Stromberg, Guo Wei, and Sanford H. Barsky
- Subjects
Cell signaling ,Stromal cell ,Angiogenesis ,Breast Neoplasms ,Mice, Transgenic ,Article ,Proto-Oncogene Protein c-ets-2 ,Malignant transformation ,Extracellular matrix ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Animals ,Humans ,PTEN ,Neoplasms, Glandular and Epithelial ,Fibroblast ,Cell Proliferation ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,biology ,PTEN Phosphohydrolase ,Mammary Neoplasms, Experimental ,Fibroblasts ,Cell cycle ,Immunity, Innate ,Extracellular Matrix ,3. Good health ,Gene Expression Regulation, Neoplastic ,Cell Transformation, Neoplastic ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Stromal Cells ,Gene Deletion - Abstract
The tumour stroma is believed to contribute to some of the most malignant characteristics of epithelial tumours. However, signalling between stromal and tumour cells is complex and remains poorly understood. Here we show that the genetic inactivation of Pten in stromal fibroblasts of mouse mammary glands accelerated the initiation, progression and malignant transformation of mammary epithelial tumours. This was associated with the massive remodelling of the extracellular matrix (ECM), innate immune cell infiltration and increased angiogenesis. Loss of Pten in stromal fibroblasts led to increased expression, phosphorylation (T72) and recruitment of Ets2 to target promoters known to be involved in these processes. Remarkably, Ets2 inactivation in Pten stroma-deleted tumours ameliorated disruption of the tumour microenvironment and was sufficient to decrease tumour growth and progression. Global gene expression profiling of mammary stromal cells identified a Pten-specific signature that was highly represented in the tumour stroma of patients with breast cancer. These findings identify the Pten-Ets2 axis as a critical stroma-specific signalling pathway that suppresses mammary epithelial tumours.
- Published
- 2009
24. Noncatalytic PTEN missense mutation predisposes to organ-selective cancer development in vivo
- Author
-
Helen M. Chamberlin, Thac Pham, Paul J. Goodfellow, Robert Pilarski, David E. Cohn, Charles L. Shapiro, Onur Egriboz, Adrian A. Suarez, Julie A. Wallace, Raleigh D. Kladney, Xing Tang, Changxian Shen, Gustavo Leone, Cynthia Timmers, Maria C. Cuitiño, Chelsea K. Martin, Kang Sup Shim, Michael C. Ostrowski, David M. O'Malley, Arunima Srivastava, Vincenzo Coppola, Thierry Pécot, Diego H. Castrillon, Morag Park, Soledad Fernandez, Hui Wang, Matthew K. Karikomi, Melissa J. Mauntel, Floor J. Backes, Thomas J. Rosol, Krista M. D. La Perle, Erin Macrae, Xiaokui Mo, Enrico Caserta, Christopher Koivisto, Sarmila Majumder, Lianbo Yu, Comprehensive Cancer Center [Colombus], Ohio State University [Columbus] (OSU), Department of Molecular Genetics [Colombus], Department of Molecular Virology, Immunology and Medical Genetics [Colombus], Ohio State University [Columbus] (OSU)-College of Medicine and Public Health [Colombus], Space-timE RePresentation, Imaging and cellular dynamics of molecular COmplexes (SERPICO), Inria Rennes – Bretagne Atlantique, Institut National de Recherche en Informatique et en Automatique (Inria)-Institut National de Recherche en Informatique et en Automatique (Inria), Center for Biostatistics [Colombus], Department of Biomedical Informatics [Columbus], Department of Biochemistry [Montréal], McGill University = Université McGill [Montréal, Canada], Rosalind and Morris Goodman Cancer Center [Montreal], Department of Oncology [Montréal], Department of Veterinary Biosciences [Colombus], College of Veterinary Medicine [Colombus], Ohio State University [Columbus] (OSU)-Ohio State University [Columbus] (OSU), Department of Pathology [Dallas], University of Texas Southwestern Medical Center [Dallas], Department of Obstetrics and Gynecology [Colombus], Department of Pathology [Columbus], and Department of Internal Medicine [Colombus]
- Subjects
Phosphatase ,Mutation, Missense ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Mice ,Genetics ,Carcinoma ,medicine ,PTEN ,Missense mutation ,Tensin ,Animals ,Gene Knock-In Techniques ,Protein kinase B ,Cells, Cultured ,C2 domain ,Cell Nucleus ,biology ,Protein Stability ,PTEN Phosphohydrolase ,[SDV.BBM.BM]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Molecular biology ,medicine.disease ,Embryo, Mammalian ,Enzyme Activation ,Oncogene Protein v-akt ,[SDV.GEN.GA]Life Sciences [q-bio]/Genetics/Animal genetics ,biology.protein ,Cancer research ,Female ,Signal transduction ,Developmental Biology ,Research Paper ,Signal Transduction - Abstract
Inactivation of phosphatase and tensin homology deleted on chromosome 10 (PTEN) is linked to increased PI3K–AKT signaling, enhanced organismal growth, and cancer development. Here we generated and analyzed Pten knock-in mice harboring a C2 domain missense mutation at phenylalanine 341 (PtenFV), found in human cancer. Despite having reduced levels of PTEN protein, homozygous PtenFV/FV embryos have intact AKT signaling, develop normally, and are carried to term. Heterozygous PtenFV/+ mice develop carcinoma in the thymus, stomach, adrenal medulla, and mammary gland but not in other organs typically sensitive to Pten deficiency, including the thyroid, prostate, and uterus. Progression to carcinoma in sensitive organs ensues in the absence of overt AKT activation. Carcinoma in the uterus, a cancer-resistant organ, requires a second clonal event associated with the spontaneous activation of AKT and downstream signaling. In summary, this PTEN noncatalytic missense mutation exposes a core tumor suppressor function distinct from inhibition of canonical AKT signaling that predisposes to organ-selective cancer development in vivo.
- Published
- 2015
25. HDAC inhibitor AR-42 decreases CD44 expression and sensitizes myeloma cells to lenalidomide
- Author
-
Luciano Cascione, Jessica Consiglio, Nicola Zanesi, Yvonne A. Efebera, Lara Rizzotto, Andrew Stiff, Flavia Pichiorri, Alessandro Canella, Balveen Kaur, Enrico Caserta, John C. Byrd, Hector Cordero Nieves, Hanna S. Radomska, Xiaokui Mo, Volinia Stefano, Emily Smith, Craig C. Hofmeister, and Douglas W. Sborov
- Subjects
Gerontology ,Blotting, Western ,lenalidomide ,Mice, Nude ,Angiogenesis Inhibitors ,Apoptosis ,Enzyme-Linked Immunosorbent Assay ,Real-Time Polymerase Chain Reaction ,Phenylbutyrate ,Immunoenzyme Techniques ,Mice ,In vivo ,medicine ,Tumor Cells, Cultured ,Animals ,Humans ,RNA, Messenger ,CD44 ,Dexamethasone ,Multiple myeloma ,Lenalidomide ,Cell Proliferation ,IGF2BP3 ,business.industry ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Profiling ,medicine.disease ,Flow Cytometry ,miR-9–5p ,Phenylbutyrates ,Xenograft Model Antitumor Assays ,3. Good health ,Thalidomide ,Histone Deacetylase Inhibitors ,medicine.anatomical_structure ,Hyaluronan Receptors ,myeloma ,Oncology ,Drug Resistance, Neoplasm ,Cancer research ,Drug Therapy, Combination ,Female ,Bone marrow ,HDAC Inhibitor AR-42 ,business ,Multiple Myeloma ,medicine.drug ,Research Paper - Abstract
Multiple myeloma (MM) is a hematological malignancy of plasma cells in the bone marrow. Despite multiple treatment options, MM is inevitably associated with drug resistance and poor outcomes. Histone deacetylase inhibitors (HDACi's) are promising novel chemotherapeutics undergoing evaluation in clinical trials for the potential treatment of patients with MM. Although in preclinical studies HDACi's have proven anti-myeloma activity, but in the clinic single-agent HDACi treatments have been limited due to low tolerability. Improved clinical outcomes were reported only when HDACi's were combined with other drugs. Here, we show that a novel pan-HDACi AR-42 downregulates CD44, a glycoprotein that has been associated with lenalidomide and dexamethasone resistance in myeloma both in vitro and in vivo. We also show that this CD44 downregulation is in part mediated by miR-9-5p, targeting insulin-like growth factor 2 mRNA binding protein 3 (IGF2BP3), which directly binds to CD44 mRNA and increases its stability. Importantly, we also demonstrate that AR-42 enhances anti-myeloma activity of lenalidomide in primary MM cells isolated from lenalidomide resistant patients and in in vivo MM mouse model. Thus, our findings shed light on potential novel combinatorial therapeutic approaches modulating CD44 expression, which may help overcome lenalidomide resistance in myeloma patients.
- Published
- 2015
26. Daratumumab Impairs Myeloma Cell Adhesion Mediated Drug Resistance through CD38 Internalization
- Author
-
Amrita Krishnan, Enrico Caserta, Cesar Terrazas, Flavia Pichiorri, Abhay R. Satoskar, Domenico Viola, Jayeeta Ghose, and Craig C. Hofmeister
- Subjects
Stromal cell ,Cell growth ,business.industry ,Bortezomib ,T cell ,Immunology ,Daratumumab ,Cell Biology ,Hematology ,CD38 ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Proteasome inhibitor ,medicine ,Cancer research ,business ,Cell adhesion ,030215 immunology ,medicine.drug - Abstract
Introduction: Daratumumab (HuMax-CD38, Dara) is an immunoglobulin G1 kappa (IgG1k) human monoclonal antibody that binds a CD38 epitope that has been recently approved by the Food and Drugs Administration as a single agent for the treatment of Multiple Myeloma (MM). Multiple myeloma (MM) is a plasma cell disorder affecting approximately 83,000 US citizens with 30,330 new cases per year in the US. The discovery of intra-clonal heterogeneity strengthens the scientific rationale of using novel therapy combinations to overcome mechanisms of resistance. While CD38 participates in NAD+ hydrolysis generating adenosine and influences intracellular calcium homeostasis through cADPR and NAADP synthesis, CD38 facilitates bone marrow (BM) homing of plasma cells through interaction with CD31 which is highly expressed in MM BM stromal cells (BMSCs) and macrophages (BM-M). Since adhesion of MM cells to stromal cells induces cell adhesion mediated drug resistance (CAM-DR), in this work, tested whether CD38 internalization after Daratumumab treatment impairs stromal cell adhesion, sensitizing MM cells to other drugs including proteasome inhibitors. Methods:Flow cytometry analysis and single cell flow analysis was done to measure the extent of surface CD38 internalization into MM cells (MM cell lines and primary cells) in vitro and ex-vivo. Adhesion assays were performed using MM cell lines treated with Daratumumab and co-cultured with BMSCs and BM-M. Apoptotic assays including cell proliferation and Annexin-V/PI staining were done to assess proteasome inhibitor induced apoptosis (bortezomib, BTZ) of MM cells pretreated with Daratumumab in the presence or absence of BM stroma. Chou-Talalay synergy analysis was used to assess the ability of Daratumumab to synergize with BTZ. Results:Single cell flow analysis revealed surface CD38 internalization into MM cell lines (MM1.S, H929, L363, RPMI-8226) and in primary myeloma cells upon incubation with increasing doses of Daratumumab. Our data show that MM cell lines and primary CD138+ MM plasma cells (MM-PCs) revealed loss of adhesion in a dose and time dependent fashion in co-culture experiments with BMSc. Moreover our data indicate that both BMSCs and BM-M protect MM cells to BTZ treatments. In order to investigate whether loss of adhesion of MM cells deprives them of protection, MM cell lines and primary cells were treated with Daratumumab and co-cultured with BM stroma and then treated with BTZ. Interestingly, it was observed that although stromal cells could protect MM cells from induced apoptosis, it failed to do so when MM cells were pretreated with Daratumumab. A more than two-fold increase in MM cell apoptosis was observed with Daratumumab-BTZ combination compared to the single agent treatments. This indicates that Daratumumab potentiates BTZ killing of MM cells. Conclusion:Daratumumab in combination with both proteasome inhibitors and immune modulators (IMiDs) are synergistic as evidenced by the results of CASTOR and POLLUX trials respectively, but the mechanisms of killing and resistance will likely be different. The main anti-MM effect of Daratumumab has so far been attributed to its antibody-dependent cellular cytotoxicity, complement dependent cellular cytotoxicity, antibody-dependent cellular phagocytosis activities and in promoting T cell expansion in relapsed/refractary MM patients enrolled in Daratumumab monotherapy trials. Our data indicate that Daratumumab potentiates the BTZ killing of MM cells via CD38 internalization, providing rationale to further explore CD38 targeting using other drugs or cell therapies. Disclosures Hofmeister: Celgene: Research Funding; Karyopharm Therapeutics: Research Funding; Arno Therapeutics, Inc.: Research Funding; Incyte, Corp: Membership on an entity's Board of Directors or advisory committees; Signal Genetics, Inc.: Membership on an entity's Board of Directors or advisory committees; Janssen: Pharmaceutical Companies of Johnson & Johnson: Research Funding; Takeda Pharmaceutical Company: Research Funding; Teva: Membership on an entity's Board of Directors or advisory committees.
- Published
- 2016
27. Small RNA Deep Sequencing Highlights the Important Contribution of Mirnas in Regulating IRF4/c-Myc Axis in Myeloma Development
- Author
-
Hector Cordero, Liu Joseph, Flavia Pichiorri, Alessandro Canella, Santhanam Ramasamy, Yvonne A. Efebera, Stefano Volinia, Enrico Caserta, Cascione Luciano, Hanna S. Radomska, Consiglio Jessica, Alberto Rocci, and Craig C. Hofmeister
- Subjects
Small RNA ,Immunology ,RNA ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Molecular biology ,RNA silencing ,Gene expression ,microRNA ,medicine ,Transcriptional regulation ,Gene silencing ,Monoclonal gammopathy of undetermined significance - Abstract
Introduction: Multiple myeloma (MM) is a hematologic malignancy characterized by the accumulation of plasma cells (PCs) in the bone marrow (BM). Over 50% of patients die within 5 years of diagnosis. The transition from normal PCs to active MM, via premalignant condition (monoclonal gammopathy of undetermined significance; MGUS), consists of many oncogenic events, including upregulation of cyclin D1 and IRF4 genes, as well as activating mutations in NRAS, and KRAS. Despite recent advances in oncogenomics, further studies are needed to identify critical players in MM pathogenesis that could be targeted for pharmacological intervention to improve outcome. Recently, aberrations in epigenetic modifications, including DNA modifications and miRNA expression have been shown to play a crucial role in development and progression of MM. miRNAs are short non-coding RNAs that bind to complementary sequences on target messenger RNA (mRNAs) and downregulate their expression by inhibiting mRNA translation, or inducing its degradation. miRNA analysis, may lead to improved understanding of MM biology and classification, by establishing associations between gene expression changes and MM molecular and clinical features. To assess whether miRNA deregulation plays a critical role in the development of MM we performed small RNA next generation sequencing in the PCs isolated from 3 patients at the MGUS stage and after that they developed active disease, but still untreated. miRNA deregulation was also validated in an independent set of newly diagnosed MM patients (n.34) compared to non-cancer age matched donors (n.6). Mechanisms of transcriptional regulation and biological roles of the differentially expressed miRNAs were also analyzed. Methods: 1x106 CD138+ frozen PCs (purity>90%) from different 3 donors before and after the disease development were used for the analysis. RNA was extracted with RNA-DNA-protein kit (Norgen Biotek) and 0.8µg of total RNA was used to generate the cDNA libraries using TruSeq Small RNA Library Preparation Kit. The obtained cDNA libraries were sequenced on an ILLUMINA system through the OSU Genomic Shared Resource (GSR). Myeloma cells (MM.1S) were treated with pan-HDACi for 24 hours and total miRNA expression was analyzed by nCounter microRNA array (NanoString Technologies, Inc.). miRNA deregulation upon use of several pan-HDAC'i in clinical use (LBH589, SAHA and AR-42) were validated in 4 different cell lines and in the MM cells of newly diagnosed MM patients. Chromatin immunoprecipitation, silencing RNA for specific histone deacetylase enzymes (HDACI, HDAC2, HDAC3, and HDAC6), western blot analysis, luciferase assays, stem loop RT-PCR, q-RT-PCR and cell proliferation assays were also performed. Results: We found that several miRNAs are commonly deregulated during disease transition. Some of these miRNAs, including miR-223, miR-221, miR-222, miR-92a and miR-93 (p Conclusions: Collectively, we believe, that these findings support the key role of miRNA aberrant expression in PC transformation. Their role in regulating the expression of IRF4 during myeloma development and lead us to speculate that this explains why IRF4 and c-Myc mRNA levels are higher in newly diagnosed MM patients, without obvious chromosomal abnormalities, compared to MGUS patients. Understanding of the molecular bases of how c-Myc expression can be regulated by a specific histone deacetylase via modulation of miRNA levels will have important impact not only on myeloma therapy, but also other hematopoietic malignancies involving c-Myc deregulation. Disclosures No relevant conflicts of interest to declare.
- Published
- 2015
28. Identifying Nuclear Phenotypes Using Semi-supervised Metric Learning
- Author
-
Shantanu Singh, Jens Rittscher, Gustavo Leone, Firdaus Janoos, Raghu Machiraju, Enrico Caserta, Thierry Pécot, Broad Institute of MIT and Harvard (BROAD INSTITUTE), Harvard Medical School [Boston] (HMS)-Massachusetts Institute of Technology (MIT)-Massachusetts General Hospital [Boston], Space-timE RePresentation, Imaging and cellular dynamics of molecular COmplexes (SERPICO), Inria Rennes – Bretagne Atlantique, Institut National de Recherche en Informatique et en Automatique (Inria)-Institut National de Recherche en Informatique et en Automatique (Inria), Ohio State University [Columbus] (OSU), Institute of Biomedical Engineering [Oxford] (IBME), University of Oxford, and Department of Computer Science and Engineering [Columbus] (CSE)
- Subjects
Stromal cell ,Computer science ,Breast Neoplasms ,Computational biology ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,medicine.disease_cause ,Sensitivity and Specificity ,Article ,Pattern Recognition, Automated ,Mice ,[INFO.INFO-LG]Computer Science [cs]/Machine Learning [cs.LG] ,Artificial Intelligence ,Cell Line, Tumor ,Image Interpretation, Computer-Assisted ,medicine ,Animals ,Computer vision ,Tumor stroma ,ComputingMilieux_MISCELLANEOUS ,Cell Nucleus ,Microscopy, Confocal ,Training set ,business.industry ,Reproducibility of Results ,Image Enhancement ,Phenotype ,Cell nucleus ,medicine.anatomical_structure ,[INFO.INFO-TI]Computer Science [cs]/Image Processing [eess.IV] ,Metric (mathematics) ,Labeled data ,Artificial intelligence ,business ,Carcinogenesis ,Algorithms - Abstract
In systems-based approaches for studying processes such as cancer and development, identifying and characterizing individual cells within a tissue is the first step towards understanding the large-scale effects that emerge from the interactions between cells. To this end, nuclear morphology is an important phenotype to characterize the physiological and differentiated state of a cell. This study focuses on using nuclear morphology to identify cellular phenotypes in thick tissue sections imaged using 3D fluorescence microscopy. The limited label information, heterogeneous feature set describing a nucleus, and existence of sub-populations within cell-types makes this a difficult learning problem. To address these issues, a technique is presented to learn a distance metric from labeled data which is locally adaptive to account for heterogeneity in the data. Additionally, a label propagation technique is used to improve the quality of the learned metric by expanding the training set using unlabeled data. Results are presented on images of tumor stroma in breast cancer, where the framework is used to identify fibroblasts, macrophages and endothelial cells - three major stromal cells involved in carcinogenesis. © 2011 Springer-Verlag.
- Published
- 2011
29. Non-parametric population analysis of cellular phenotypes
- Author
-
Thierry Pécot, Enrico Caserta, Shantanu Singh, Gustavo Leone, Kun Huang, Firdaus Janoos, Raghu Machiraju, Jens Rittscher, Broad Institute of MIT and Harvard (BROAD INSTITUTE), Harvard Medical School [Boston] (HMS)-Massachusetts Institute of Technology (MIT)-Massachusetts General Hospital [Boston], Space-timE RePresentation, Imaging and cellular dynamics of molecular COmplexes (SERPICO), Inria Rennes – Bretagne Atlantique, Institut National de Recherche en Informatique et en Automatique (Inria)-Institut National de Recherche en Informatique et en Automatique (Inria), Ohio State University [Columbus] (OSU), Department of Biomedical Informatics [Columbus], Institute of Biomedical Engineering [Oxford] (IBME), University of Oxford, and Department of Computer Science and Engineering [Columbus] (CSE)
- Subjects
Population ,Cell ,Cytological Techniques ,Genomics ,Breast Neoplasms ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Computational biology ,Disease ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Biology ,Bioinformatics ,Article ,Mice ,medicine ,Image Processing, Computer-Assisted ,Animals ,Humans ,education ,skin and connective tissue diseases ,Gene ,ComputingMilieux_MISCELLANEOUS ,Cell Nucleus ,education.field_of_study ,Microscopy ,Nonparametric statistics ,PTEN Phosphohydrolase ,Cell Biology ,Fibroblasts ,Models, Theoretical ,Phenotype ,Cell nucleus ,medicine.anatomical_structure ,[INFO.INFO-TI]Computer Science [cs]/Image Processing [eess.IV] ,Female ,sense organs ,Algorithms - Abstract
Methods to quantify cellular-level phenotypic differences between genetic groups are a key tool in genomics research. In disease processes such as cancer, phenotypic changes at the cellular level frequently manifest in the modification of cell population profiles. These changes are hard to detect due the ambiguity in identifying distinct cell phenotypes within a population. We present a methodology which enables the detection of such changes by generating a phenotypic signature of cell populations in a data-derived feature-space. Further, this signature is used to estimate a model for the redistribution of phenotypes that was induced by the genetic change. Results are presented on an experiment involving deletion of a tumor-suppressor gene dominant in breast cancer, where the methodology is used to detect changes in nuclear morphology between control and knockout groups. © 2011 Springer-Verlag.
- Published
- 2011
30. Analysis of spatial variation of nuclear morphology in tissue microenvironments
- Author
-
Shantanu Singh, Gustavo Leone, Michael C. Ostrowski, Sundaresan Raman, Enrico Caserta, Jens Rittscher, and Raghu Machiraju
- Subjects
Pathology ,medicine.medical_specialty ,Tumor microenvironment ,Stromal cell ,Cell ,Locus (genetics) ,Biology ,Cell morphology ,medicine.disease ,Phenotype ,Cell biology ,medicine.anatomical_structure ,Breast cancer ,Tumor progression ,medicine - Abstract
We present a study of the spatial variation of nuclear morphology of stromal and cancer-associated fibroblasts in the mouse mammary gland. The work is part of a framework being developed for the analysis of the tumor microenvironment in breast cancer. Recent research has uncovered the role of stromal cells in promoting tumor growth and progression. In specific, studies have indicated that stromal fibroblasts - formerly considered to be passive entities in the extra-cellular matrix - play an active role in the progression of tumor in mammary tissue. We have focused on the analysis of the nuclear morphology of fibroblasts, which several studies have shown to be a critical phenotype in cancer. An essential component of our approach is that the nuclear morphology is studied within the 3D spatial context of the tissue, thus enabling us to pose questions about how the locus of a cell relates to its morphology, and possibly to its function. In order to make quantitative comparisons between nuclear populations, we build statistical shape models of cell populations and infer differences between the populations through these models. We present our observation on both normal and tumor tissues from the mouse mammary gland. ©2010 IEEE.
- Published
- 2010
31. Abstract 3260: Allele-specific tumor spectrum in Pten knockin mice
- Author
-
Matt Karikomi, Thomas J. Rosol, Enrico Caserta, Hui Wang, Michael C. Ostrowski, Hui-Zi Chen, Paul C. Stromberg, Gustavo Leone, Michael Weinstein, Krista M. D. La Perle, Julie Moffitt, Ravi Rajmohan, Soledad Fernandez, Maysoon Rawahneh, Charis Eng, Shan Naidu, and Julie A. Stephens
- Subjects
Genetics ,Cancer Research ,biology ,business.industry ,Cancer ,medicine.disease ,Phenotype ,law.invention ,Germline mutation ,Oncology ,law ,Lipid phosphatase activity ,Cancer research ,biology.protein ,Medicine ,Suppressor ,Missense mutation ,PTEN ,Allele ,business - Abstract
Germline mutations in the PTEN tumor suppressor cause Cowden and Bannayan-Riley-Ruvalcaba syndromes, two dominantly inherited disorders characterized by mental retardation, multiple hamartomas and variable cancer risk. Here we modeled three sentinel mutant alleles of PTEN identified in Cowden patients and show that the nonsense PtenΔ4–5 and missense PtenC124R and PtenG129E alleles lacking lipid phosphatase activity cause similar developmental abnormalities but distinct tumor spectra with varying severity and age-of-onset. Allele-specific differences may be accounted by loss-of-function for Pten Δ4-5, hypomorphic function for PtenC124R and gain-of-function for PtenG129E. These data demonstrate that the variable tumor phenotypes observed in Cowden and Bannayan-Riley-Ruvalcaba syndrome patients can be attributed to specific mutations in PTEN that alter protein function through distinct mechanisms. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 3260.
- Published
- 2010
32. Interleukin-15 Is a Proto-Oncogene In Acute Large Granular Lymphocyte Leukemia
- Author
-
Pierluigi Porcu, Nyla A. Heerema, Michael A. Caligiuri, Xunchang Zou, Heiko Becker, Laura Sullivan, Harold A. Fisk, Ramasamy Santhanam, Lauren G. Falkenberg, Lai-Chu Wu, Laura Jaroncyk, Deanna M. W. Schaefer, Ramiro Garzon, Anjali Mishra, Sebastian Schwind, Shujun Liu, Jadwiga Labanowska, Laura J. Rush, Guido Marcucci, Douglas P. Curphey, Enrico Caserta, Gregory H. Sams, Jason C. Chandler, and Yue-Zhong Wu
- Subjects
Oncogene ,medicine.medical_treatment ,Immunology ,Wild type ,Aneuploidy ,Interleukin ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Molecular biology ,Cytokine ,Interleukin 15 ,In vivo ,Splenocyte ,medicine - Abstract
Abstract 704 Interleukin (IL)-15 is critical for the differentiation, proliferation, activation and survival of large granular lymphocytes (LGL). Malignant blasts from patients with acute LGL leukemia (LGLL) can express membrane bound IL-15 and often require IL-15 or IL-2 to survive and expand in vitro, suggesting a pivotal role of IL-15 in the genesis of LGLL in vivo. Indeed, 30% of mice engineered to over express IL-15 develop LGLL (J Exp Med 193:219-231, 2001), suggesting that IL-15 is a proto-oncogene. The present study examined the mechanism by which this may occur in mouse and in man. We observed ~2.5-fold increased levels of DNA methyltransferase 3b (Dnmt3b) in IL-15tg mice with LGLL compared to wild type (Wt) splenocytes (2.6 ± 0.6 -fold higher, N = 3 each, P =.03) and a ~2-fold increased levels of global DNA methylation (GDM) compared to Wt splenocytes (% global DNA levels measured by mass spec as % 5mC/(5mC+2dC): 3.6 ± 0.11%, N = 4 for IL-15tg LGLL; 1.5 ± 0.08%, N = 4 for Wt, P Disclosures: No relevant conflicts of interest to declare.
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.