13 results on '"Rin Nakamura"'
Search Results
2. Pretreatment with Tocilizumab Prior to the CD3 Bispecific Cevostamab in Patients with Relapsed/Refractory Multiple Myeloma (RRMM) Showed a Marked Reduction in Cytokine Release Syndrome Incidence and Severity
- Author
-
Suzanne Trudel, Nizar J. Bahlis, Andrew Spencer, Rayan Kaedbey, Paula Rodriguez Otero, Simon J Harrison, Chihunt Wong, Grant R. Goodman, Rin Nakamura, Voleak Choeurng, James Cooper, and Maria-Victoria Mateos
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
3. A BCMA/CD16A bispecific innate cell engager for the treatment of multiple myeloma
- Author
-
Satoko Kakiuchi-Kiyota, Thorsten Ross, Heidi Ackerly Wallweber, James R. Kiefer, Melissa M. Schutten, Adeyemi O. Adedeji, Hao Cai, Robert Hendricks, Sivan Cohen, Srividya Myneni, Luna Liu, Aaron Fullerton, Nicholas Corr, Lanlan Yu, Denise de Almeida Nagata, Shelly Zhong, Steven R. Leong, Ji Li, Rin Nakamura, Teiko Sumiyoshi, Jinze Li, Ayse Meric Ovacik, Bing Zheng, Mike Dillon, Christoph Spiess, Susanne Wingert, Erich Rajkovic, Kristina Ellwanger, Uwe Reusch, and Andrew G. Polson
- Subjects
Cancer Research ,Oncology ,Phagocytosis ,Antibodies, Bispecific ,Receptors, IgG ,Humans ,Hematology ,B-Cell Maturation Antigen ,Multiple Myeloma - Abstract
Despite the recent progress, multiple myeloma (MM) is still essentially incurable and there is a need for additional effective treatments with good tolerability. RO7297089 is a novel bispecific BCMA/CD16A-directed innate cell engager (ICE
- Published
- 2021
4. Twenty-four-hour continuous and remote monitoring of respiratory rate using a medical radar system for the early detection of pneumonia in symptomatic elderly bedridden hospitalized patients
- Author
-
Masakazu Okada, Taro Matsuo, Yukiya Hakozaki, Guanghao Sun, Takemi Matsui, Tetsuo Kirimoto, and Rin Nakamura
- Subjects
medicine.medical_specialty ,medical radar ,Medical staff ,Respiratory rate ,Hospitalized patients ,business.industry ,Early detection ,Case Report ,respiratory rate ,Workload ,Case Reports ,General Medicine ,030204 cardiovascular system & hematology ,medicine.disease ,Radar systems ,03 medical and health sciences ,Pneumonia ,0302 clinical medicine ,Quality of life ,030220 oncology & carcinogenesis ,Emergency medicine ,medicine ,pneumonia ,long‐term monitoring ,business - Abstract
The use of continuous and long‐term monitoring of respiratory rate is vital for predicting pneumonia in symptomatic patients; however, it is often measured manually and discontinuously by counting of chest wall movements in routine practice.1 Hence, here we developed a point‐of‐care system for the early detection of pneumonia in symptomatic elderly bedridden hospitalized patients on the basis of 24‐hours continuous and noncontact monitoring of respiratory rate using a medical radar sensor. We focused on designing a system that would improve hospitalized patient quality of life and reduce medical staff workload. To this end, we adopted a medical radar sensor for respiration monitoring that featured an extremely low burden on the patient and enabled unobtrusive measurements without the need to attach electrodes to the patient's body.2 To reduce medical staff workload, a prediction method, that is, return map,3 was implemented into the system to analyze the time series respiratory rates, thereby extracting the risk period of pneumonia and sending out an alarm. Infection is the most common cause of hospital‐acquired pneumonia, which is associated with prolonged hospital stay and increased mortality in elderly hospitalized patients.4 In this study, we evaluated the feasibility of this system for detecting the risk period of pneumonia on the basis of continuously monitored respiratory rates in elderly bedridden hospitalized patients.
- Published
- 2018
- Full Text
- View/download PDF
5. Early Pharmacodynamic Changes in T-Cell Activation, Proliferation, and Cytokine Production Confirm the Mode of Action of BFCR4350A, a FcRH5/CD3 T-Cell-Engaging Bispecific Antibody, in Patients with Relapsed/Refractory Multiple Myeloma
- Author
-
Deanna Grant Wilson, James R. Cooper, Teiko Sumiyoshi, Sean Lear, Hartmut Koeppen, Adam D. Cohen, Anjali Vaze, Simon J. Harrison, Rin Nakamura, Andrew Spencer, Suzanne Trudel, Mengsong Li, and Bernard M. Fine
- Subjects
Oncology ,medicine.medical_specialty ,Bispecific antibody ,Immunological synapse formation ,business.industry ,medicine.medical_treatment ,T cell ,Immunology ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Cytokine ,medicine.anatomical_structure ,Pharmacodynamics ,Internal medicine ,Relapsed refractory ,Medicine ,In patient ,business ,Multiple myeloma - Abstract
Introduction: Fc receptor-homolog 5 (FcRH5) is an immunoglobulin (Ig) domain-containing type I membrane protein that is expressed exclusively in the B-cell lineage. FcRH5 expression is retained in myeloma cells, with near 100% prevalence, and is elevated vs normal B cells. BFCR4350A is a humanized IgG-based T-cell-engaging bispecific antibody. Binding of BFCR4350A to the most membrane-proximal domain of FcRH5 on myeloma cells and to cluster of differentiation 3 (CD3) on T cells results in efficient immunological synapse formation and potent T-cell-directed killing of myeloma cells (Li, et al. Cancer Cell 2017). An ongoing Phase I dose-escalation study (GO39775; NCT03275103) is investigating the safety, activity, pharmacodynamics (PD) and pharmacokinetics of BFCR4350A monotherapy in patients (pts) with relapsed/refractory (R/R) multiple myeloma (MM). In Arm A, clinical activity was observed at the 3.6mg/20mg (step/target) dose level and above, and toxicity was manageable (Cohen, et al. ASH 2020). We present preliminary biomarker data that demonstrate the mode of action (MOA) of BFCR4350A, provide support for Cycle (C) 1 step-up dosing, and offer preliminary insights into markers that may predict response. Methods: In Arm A, R/R MM pts receive BFCR4350A by intravenous infusion in 21-day cycles. In C1, a single step-up dosing approach is used to mitigate the risk for cytokine release syndrome (CRS), with the step dose given on C1 Day (D) 1 and the target dose given on C1D8. The target dose is then administered on D1 of each subsequent cycle. PD changes in peripheral blood (PB) are assessed at baseline and at multiple time points within C1 by whole blood flow cytometry, plasma cytokine electrochemiluminescence and digital ELISA. Tumor biomarkers are assessed at baseline and pre-C2 by bone marrow (BM) biopsy dual CD138/CD8 immunohistochemistry staining and BM aspirate flow cytometry. The clinical cut-off date used for the current analyses was April 13, 2020. Results: At cut-off, all pts in Arm A (n=51) were biomarker evaluable. FcRH5 expression on myeloma cells was detected in all pts. Dose-dependent PD changes in PB were observed 24-192 hrs after the C1D1 infusion. Variable reduction in circulating T cells was observed 24 hrs after the 0.3-1.8mg C1D1 doses, while consistent reduction was observed after the 3.6mg C1D1 dose, with recovery by C1D8 (192 hrs). T-cell activation was detected 24 hrs post-infusion by upregulation of CD69 in CD8 and CD4 T cells and elevation of IFN-γ in plasma (median increase of ~150-fold from baseline), while T-cell proliferation (Ki67+) peaked by C1D8. At the 3.6mg C1D1 dose, CD8 T-cell activation and proliferation were up to 20-fold higher than at baseline. Minimal elevation of IL-6 was observed post-infusion in pts who received doses below 3.6mg on C1D1, while more consistent increases (≥100pg/ml) were observed in pts who received 3.6mg. IL-6 levels peaked within 24 hrs of the C1D1 dose and the kinetics of IL-6 increase were associated with dose and risk for CRS. Step-up dosing mitigated the risk for severe CRS, as evidenced by lower IL-6 levels after the C1D8 target dose compared to the 3.6mg C1D1 step dose in 27/33 (82%) pts (see Cohen, et al. ASH 2020 for corresponding clinical data). Preliminary data suggest that pts who respond to BFCR4350A have more pronounced T-cell expansion in PB, irrespective of baseline CD8 T-cell levels during the first week of C1. Analysis of the subset of pts with paired BM biopsies (n=19 pts) revealed that levels of CD8+ tumor infiltrating T-cells (TILs) were higher in responding pts than in non-responding pts at the end of C1. Conclusions: In this study, we demonstrated that early PD changes in T-cell activation, proliferation, and cytokine production confirm the MOA of BFCR4350A and support C1 step-up dosing for CRS mitigation in R/R MM. Early data suggest that at the end of C1, higher peripheral CD8 T-cell expansion and TILs are observed in responding pts than in non-responding pts. Additional analyses are ongoing to establish BFCR4350A response predictors and to better characterize the populations that are likely to benefit. Updated data will be presented. Disclosures Nakamura: Genentech, Inc.: Current Employment; F. Hoffmann-La Roche: Current equity holder in publicly-traded company. Lear:F. Hoffmann-La Roche: Current equity holder in publicly-traded company; Genentech, Inc.: Current Employment. Wilson:Genentech, Inc.: Current Employment. Koeppen:Genentech, Inc./ F. Hoffmann-La Roche: Current Employment; F. Hoffmann-La Roche, Pliant Therapeutics, Allogene, Jounce: Current equity holder in publicly-traded company. Vaze:Genentech, Inc./ F. Hoffmann-La Roche: Current Employment, Current equity holder in publicly-traded company. Trudel:Takeda: Honoraria; Sanofi: Honoraria; GSK: Consultancy, Honoraria, Research Funding; Genentech, Inc.: Research Funding; BMS: Consultancy, Honoraria, Research Funding; Karyopharm: Honoraria; AstraZeneca: Honoraria; Pfizer: Honoraria, Research Funding; Amgen: Consultancy, Research Funding; Janssen: Honoraria, Research Funding. Spencer:Amgen, Celgene, Haemalogix, Janssen, Servier and Takeda: Research Funding; AbbVie, Amgen, Celgene, Haemalogix, Janssen, Sanofi, SecuraBio, Specialised Therapeutics Australia, Servier and Takeda: Honoraria; AbbVie, Celgene, Haemalogix, Janssen, Sanofi, SecuraBio, Specialised Therapeutics Australia, Servier and Takeda: Consultancy; Celgene, Janssen and Takeda: Speakers Bureau. Harrison:Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; GSK: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; F. Hoffmann-La Roche: Consultancy, Honoraria; Novartis: Consultancy, Honoraria, Patents & Royalties: wrt panobinostat; Janssen: Honoraria; BMS: Consultancy, Honoraria; CRISPR Therapeutics: Consultancy, Honoraria; Haemalogix: Consultancy; Takeda: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen-Cilag: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; AbbVie: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Amgen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Cohen:Novartis: Other: Patents/Intellectual property licensed, Research Funding; Bristol-Myers Squibb: Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Membership on an entity's Board of Directors or advisory committees; Takeda,: Membership on an entity's Board of Directors or advisory committees; Janssen: Membership on an entity's Board of Directors or advisory committees; GlaxoSmithKline: Membership on an entity's Board of Directors or advisory committees; Kite Pharma: Membership on an entity's Board of Directors or advisory committees; Oncopeptides: Membership on an entity's Board of Directors or advisory committees; Seattle Genetics: Membership on an entity's Board of Directors or advisory committees; AstraZeneca: Membership on an entity's Board of Directors or advisory committees; Genentech/Roche: Membership on an entity's Board of Directors or advisory committees. Fine:Genentech, Inc.: Current Employment; F. Hoffmann-La Roche: Current equity holder in publicly-traded company. Li:Genentech, Inc./ F. Hoffmann-La Roche: Current Employment. Cooper:Genentech, Inc./ F. Hoffmann-La Roche: Current Employment, Current equity holder in publicly-traded company. Sumiyoshi:Genentech, Inc.: Current Employment, Current equity holder in publicly-traded company, Divested equity in a private or publicly-traded company in the past 24 months. OffLabel Disclosure: BFCR4350A is a humanized IgG-based T-cell-engaging bispecific antibody that targets the most membrane-proximal domain of FcRH5 on myeloma cells and CD3 on T cells. Dual binding facilitates efficient immunological synapse formation, resulting in T-cell activation and killing of myeloma cells. BFCR4350A is an investigational agent.
- Published
- 2020
- Full Text
- View/download PDF
6. Draft genome of the globally widespread and invasive Argentine ant (Linepithema humile)
- Author
-
Jo Anne Holley, Kaitlyn A. Mathis, Marie-Julie Favé, Reed M. Johnson, Richard Benton, Abderrahman Khila, Joseph G. Laird, Juergen Gadau, Carson Holt, Martin Helmkampf, Vincent Croset, Elizabeth Cash, Eran Elhaik, Aleksey V. Zimin, Kirk J. Grubbs, Ellen van Wilgenburg, Mark Yandell, Ehab Abouheif, Jennifer E. Placek, Brian R. Johnson, Hugh M. Robertson, Rick P. Overson, Joseph A. Moeller, Hao Hu, Chris Smith, Garret Suen, Kimberly K. O. Walden, Christine G. Elsik, Elissa L. Suhr, Darren E. Hagen, Dan Graur, Cameron R. Currie, Shu Tao, Rin Nakamura, Jay W. Kim, Monica Munoz-Torres, Justin T. Reese, Joshua D. Gibson, Lumi Viljakainen, Alexander L. Wild, Candice W. Torres, Ana Sofia Ibarraran Viniegra, Rajendhran Rajakumar, James A. Yorke, Vilaiwan M. Fernandes, Marguerite C. Murphy, Andrew V. Suarez, Neil D. Tsutsui, Christopher D. Smith, and Surabhi Nigam
- Subjects
Genome, Insect ,Molecular Sequence Data ,Hierarchy, Social ,Receptors, Odorant ,Polymorphism, Single Nucleotide ,Genome ,California ,DNA sequencing ,Commentaries ,Argentine ant ,Animals ,Gene ,Phylogeny ,Illumina dye sequencing ,Gene Library ,Genetics ,Whole genome sequencing ,Multidisciplinary ,Base Sequence ,biology ,Ants ,Genomics ,Sequence Analysis, DNA ,DNA Methylation ,Biological Sciences ,biology.organism_classification ,Genetics, Population ,DNA methylation ,Linepithema - Abstract
Ants are some of the most abundant and familiar animals on Earth, and they play vital roles in most terrestrial ecosystems. Although all ants are eusocial, and display a variety of complex and fascinating behaviors, few genomic resources exist for them. Here, we report the draft genome sequence of a particularly widespread and well-studied species, the invasive Argentine ant ( Linepithema humile ), which was accomplished using a combination of 454 (Roche) and Illumina sequencing and community-based funding rather than federal grant support. Manual annotation of >1,000 genes from a variety of different gene families and functional classes reveals unique features of the Argentine ant's biology, as well as similarities to Apis mellifera and Nasonia vitripennis . Distinctive features of the Argentine ant genome include remarkable expansions of gustatory (116 genes) and odorant receptors (367 genes), an abundance of cytochrome P450 genes (>110), lineage-specific expansions of yellow/major royal jelly proteins and desaturases, and complete CpG DNA methylation and RNAi toolkits. The Argentine ant genome contains fewer immune genes than Drosophila and Tribolium , which may reflect the prominent role played by behavioral and chemical suppression of pathogens. Analysis of the ratio of observed to expected CpG nucleotides for genes in the reproductive development and apoptosis pathways suggests higher levels of methylation than in the genome overall. The resources provided by this genome sequence will offer an abundance of tools for researchers seeking to illuminate the fascinating biology of this emerging model organism.
- Published
- 2016
7. Myeloid cell biology and inhibition of anti-tumor immune responses by MPDL3280A in urothelial bladder cancer
- Author
-
Zachary Boyd, Rin Nakamura, Gregg Fine, Thomas Powles, Priti S. Hegde, Xiaodong Shen, Daniel P. Petrylak, Nicholas J. Vogelzang, Daniel S. Chen, Teiko Sumiyoshi, Christina Rabe, Mitchell Denker, Marcin Kowanetz, Siew-leng Melinda Teng, Qun J Wu, Yuanyuan Xiao, and Yohann Loriot
- Subjects
Pharmacology ,Cancer Research ,education.field_of_study ,Myeloid ,Bladder cancer ,business.industry ,Immunogenicity ,Immunology ,Population ,Stem cell marker ,medicine.disease ,Granzyme B ,Immune system ,medicine.anatomical_structure ,Oncology ,Poster Presentation ,medicine ,Cancer research ,Granzyme A ,Molecular Medicine ,Immunology and Allergy ,business ,education - Abstract
Treatment options for metastatic urothelial bladder cancer (UBC) are limited. Mutational complexity is known to be high in UBC and may correlate with increased immunogenicity. MPDL3280A, a human PD-L1 monoclonal antibody containing an engineered Fc-domain designed to promote a Th1-driven response, has demonstrated a RECIST response rate of 43% in diagnostically selected, pretreated patients with UBC. A total of 68 patients (67 with efficacy evaluable) were enrolled in the UBC cohort of the Phase I study; 45% were PD-L1 IHC diagnostic positive as defined by expression of PD-L1 on ≥ 5% of tumor-infiltrating immune cells. In the prescreened UBC population, the prevalence of PD-L1-positive patients was 27%. Comprehensive gene expression analyses of UBC tumors were conducted to interrogate the tumor immune microenvironment in PD-L1-positive tumors and to identify potential mechanisms associated with response or resistance to MPDL3280A. In this study, PD-L1-positive tumors exhibited a high prevalence of gene expression markers associated with T-effector cells (Teff), including perforin, IFNγ, CD8A, granzyme B, granzyme A and EOMES. Additionally, a low baseline signature of genes associated with myeloid cell markers, including IL1B and IL8, appeared to be statistically significantly associated (P
- Published
- 2014
- Full Text
- View/download PDF
8. Tigit, CD226 and PD-L1/PD-1 Are Highly Expressed By Marrow-Infiltrating T Cells in Patients with Multiple Myeloma
- Author
-
Jenny Wu, Andrew Glibicky, Woodard Joseph Paul, Jeffrey M. Venstrom, Connie Ma, Joanne I. Adamkewicz, Teiko Sumiyoshi, Yu-Waye Chu, Jane L. Grogan, Mahesh Yadav, Alberto Robert, John Byon, Rin Nakamura, Ray Meng, and Cherie Green
- Subjects
0301 basic medicine ,biology ,business.industry ,CD226 ,CD3 ,Immunology ,CD28 ,Cell Biology ,Hematology ,CD38 ,Biochemistry ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,TIGIT ,030220 oncology & carcinogenesis ,PD-L1 ,biology.protein ,Cancer research ,Medicine ,business ,CD8 - Abstract
Introduction:TIGIT (T-cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif [ITIM] domain) is an inhibitory immunoreceptor expressed by T and natural killer (NK) cells that is an important regulator of anti-tumor and anti-viral immunity. TIGIT shares its high-affinity ligand PVR (CD155) with the activating receptor CD226 (DNAM-1). We have recently shown that TIGIT blockade, together with PD-L1/PD-1 blockade, provides robust efficacy in syngeneic tumor and chronic viral infection models. Importantly, CD226 blockade abrogates the benefit of TIGIT blockade, suggesting additional benefit of TIGIT blockade through elaboration of CD226-mediated anti-tumor immunity, analogous to CTLA-4/CD28 regulation of T-cell immunity. Whether TIGIT and CD226 are expressed in patients with multiple myeloma (MM) and how TIGIT expression relates to PD-L1/PD-1 expression is unknown. Here we evaluate expression of TIGIT, CD226, PD-1 and PD-L1 in patients with MM to inform novel immunotherapy combinations. Methods:We performed multi-color flow cytometry (n = 25 patients), and multiplex qRT-PCR (n = 7) on bone marrow specimens from patients with MM to assess expression of TIGIT, CD226, PD-1, and PD-L1 on tumor and immune cells. Cells were stained with fluorescently conjugated monoclonal antibodies to label T cells (CD3, CD4, CD8), NK cells (CD56, CD3), plasma cells (CD38, CD45, CD319, CD56), inhibitory/activating receptors (PD-1, TIGIT, PD-L1, CD226), and an amine-reactive viability dye (7-AAD). Stained and fixed cells were analyzed by flow cytometry using BD FACSCanto™ and BD LSRFortessa™. Results:TIGIT, CD226 and PD-L1/PD-1 were detectable by flow cytometry in all patients with MM who were tested, with some overlapping and distinct expression patterns. TIGIT was commonly expressed by marrow-infiltrating CD8+ T cells (median, 65% of cells), CD4+ T cells (median, 12%) and NK cells. In contrast, CD226 was more commonly expressed by marrow-infiltrating CD4+ T cells (median, 74%) compared with CD8+ T cells (median, 38%). PD-1 was expressed by marrow-infiltrating CD8+ T cells (median 38%) and CD4+ T cells (median, 16%). TIGIT was co-expressed with PD-1 on CD8+ T cells (67%-97% TIGIT+ among PD-1+), although many PD-1-negative CD8+ T cells also expressed TIGIT (39%-78% of PD-1-negative). PD-L1 was also expressed by CD8+ (median, 23%) and CD4+ (median, 8%) T cells in addition to MM plasma cells (median, 95%), albeit with significantly lower intensity on T cells compared with plasma cells. The expression of TIGIT and PD-L1 mRNA was highly correlated (R2 = 0.80). Analysis of PVR expression will also be presented. Conclusions: TIGIT, CD226, PD-1, and PD-L1 were commonly expressed in MM bone marrow, but with different patterns. Among CD8+ T cells, the frequency of TIGIT+ T cells was almost twice that of PD-1+ T cells, whereas the majority of CD4+ T cells expressed CD226. TIGIT blockade may complement anti-PD-L1/PD-1 immunotherapy by activating distinct T-cell/NK-cell subsets with synergistic clinical benefit. These results provide new insight into the immune microenvironment of MM and rationale for targeting both the PD-L1/PD-1 interaction and TIGIT in MM. Disclosures Yadav: Genentech, Inc.: Employment. Green:Genentech, Inc.: Employment. Ma:Genentech, Inc.: Employment. Robert:Genentech, Inc.: Employment. Glibicky:Makro Technologies Inc.: Employment; Genentech, Inc.: Consultancy. Nakamura:Genentech, Inc.: Employment. Sumiyoshi:Genentech, Inc.: Employment. Meng:Genentech, Inc.: Employment, Equity Ownership. Chu:Genentech Inc.: Employment. Wu:Genentech: Employment. Byon:Genentech, Inc.: Employment. Woodard:Genentech, Inc.: Employment. Adamkewicz:Genentech, Inc.: Employment. Grogan:Genentech, Inc.: Employment. Venstrom:Roche-Genentech: Employment.
- Published
- 2016
- Full Text
- View/download PDF
9. Anti-FcRH5/CD3 T Cell Dependent Bispecific Antibody (TDB) for the Treatment of Multiple Myeloma
- Author
-
Vanessa Clark, Isidro Hötzel, Jennifer Johnston, Siddharth Sukumaran, Dionysos Slaga, Teiko Sumiyoshi, Genee Lee, Sam A. Menzies, McCarty Luke, Rin Nakamura, Elizabeth Luis, John R. James, Teemu T. Junttila, Dimitry M. Danilenko, Ji Li, Danielle DiCara, Klara Totpal, Nicola J. Stagg, Diego Ellerman, Zhengmao Ye, Ryan Cook, and Michael J. Harris
- Subjects
biology ,business.industry ,CD3 ,T cell ,Immunology ,T-cell receptor ,Cell Biology ,Hematology ,Biochemistry ,Epitope ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer cell ,biology.protein ,Cytotoxic T cell ,Medicine ,Antibody ,business ,B cell ,030215 immunology - Abstract
Bispecific antibodies that retarget cytotoxic T cell activity to kill cancer cells are currently under clinical evaluation. However, the molecular mechanism for how CD3-bispecific antibodies 'trigger' intracellular T cell signaling is not known. We demonstrate that bispecific antibodies invoke an equivalent biophysical mechanism of TCR triggering as that observed for the TCR/pMHC interaction, including target clustering and exclusion of CD45 phosphatase from the synapse. The dimensions of the target molecule play a key role in the efficiency of the synapse formation. However, we demonstrate that rational epitope selection can overcome the spatial inhibition caused by target molecules with a large extracellular domain and result in efficient synapse formation and highly potent T cell triggering. With this insight, we developed a novel T-cell dependent bispecific (TDB) antibody, anti-FcRH5/CD3 TDB, targeting the B cell lineage marker FcRH5 for multiple myeloma. Anti-FcRH5/CD3 TDB demonstrated cytotoxicity against human plasma cells and patient derived myeloma tumor cells at picomolar doses. Very low target expression level is sufficient to induce anti-FcRH5/CD3 TDB mediated killing, indicating broad activity in multiple myeloma where the prevalence of FcRH5 expression is 100%. In primates, anti-FcRH5/CD3 treatment resulted in complete depletion of tissue B cells and bone marrow plasma cells. Anti-FcRH5/CD3 TDB induces immunosuppressive feedback signaling, including PD1 up-regulation, which can be overcome by PD-L1 antibodies. These data demonstrate the potential for the anti-FcRH5/CD3 TDB, alone or in combination with inhibition of PD1/PDL1 signaling in the treatment of multiple myeloma and other B-cell malignancies. Disclosures Li: Genentech: Employment. Stagg:Genentech: Employment. Johnston:Genentech: Employment. DiCara:Genentech: Employment. Clark:Genentech: Employment. Cook:Genentech: Employment. Slaga:Genentech: Employment. Nakamura:Genentech: Employment. Luke:Genentech: Employment. Sukumaran:Genentech: Employment. Luis:Genentech: Employment. Ye:Genentech: Employment. Sumiyoshi:Genentech: Employment. Danilenko:Genentech: Employment. Lee:Genentech: Employment. Totpal:Genentech: Employment. Ellerman:Genentech: Employment. Hötzel:Genentech: Employment. Junttila:Genentech: Employment.
- Published
- 2016
- Full Text
- View/download PDF
10. Abstract 4257: Gene expression and genomic drift comparative analysis between patient-derived conditionally reprogrammed cells and original tumors
- Author
-
Timothy R. Wilson, Teiko Sumiyoshi, Mark R. Lackner, Heidi Savage, Shih-Min A. Huang, Walter C. Darbonne, Shoji Ikeda, Bonnie Liu, Jessica Li, Lukas C. Amler, Garret Hampton, and Rin Nakamura
- Subjects
Genetics ,Cancer Research ,Biology ,medicine.disease ,Primary tumor ,Gene expression profiling ,Transcriptome ,Prostate cancer ,Breast cancer ,Oncology ,Gene expression ,medicine ,Cancer research ,Gene ,Ex vivo - Abstract
First two authors contributed equally Last two authors contributed equally Recent studies have shown that ex vivo propagation of normal tissues or patient-derived tumor cells in presence of irradiated fibroblast feeder cells and ROCK inhibitor can rapidly establish conditionally reprogramed cells (CRCs). In case of normal tissues, the induction of CRCs was reversible when the ROCK inhibitor and the feeder cells were removed, resulting in CRCs differentiating to its tissue origin (Liu et al.2012). Previous publications suggested that the establishment of such cell models provides new strategies to understand acquired resistance during treatment (Crystal et al 2015) and to predict treatment response (Liu et al. 2014). However, gene expression modulations and genomic drifting during the establishment of CRC propagation have not been thoroughly studied. The primary goal of this study is to molecularly characterize alterations between the original tumor tissues and the derived models growing with or without ROCK inhibitor. Understanding in-depth molecular fluctuations in this patient-derived ex vivo system will facilitate its appropriate use for tumor biology experimentation. Herein, tumors from prostate cancer and breast cancer patients were surgical removed and cryopreserved at the clinical site then processed and cultured as previously described (Liu et al. 2012). Gene expression profiling and next-generation sequencing were carried out on the original tumor tissues and cellular models passaged during the CRC propagation in the presence or absence of ROCK inhibitor. Gene expression analysis of the prostate cancer cells and the breast cancer cells were carried out using a 93-gene prostate cancer-focused Fluidigm panel and a 800 gene NanoString breast cancer-focused panel, respectively. Cancer hotspot mutations were analyzed using the Ion Torrent Cancer Hotspot v2 NGS assay. Through aforementioned genomic and transcriptomic interrogations, we demonstrated the extent of indication-relevant gene expression modulation during establishment and propagation of these cells. We also characterize cancer hotspot mutations in the primary tumor cells and the stability of those mutations during ex vivo propagation. These results should begin to inform the appropriate use of the CRC model for tumor biology experimentation. Citation Format: Jessica Li, Bonnie Liu, Rin Nakamura, Heidi Savage, Shoji Ikeda, Timothy Wilson, Teiko Sumiyoshi, Garret Hampton, Lukas Amler, Mark Lackner, Shih-Min A. Huang, Walter C. Darbonne. Gene expression and genomic drift comparative analysis between patient-derived conditionally reprogrammed cells and original tumors. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4257.
- Published
- 2016
- Full Text
- View/download PDF
11. Draft genome of the red harvester ant Pogonomyrmex barbatus
- Author
-
Monica Munoz-Torres, Vincent Croset, Reed M. Johnson, Jürgen Gadau, Richard Benton, Abderrahman Khila, Surabhi Nigam, Martin Helmkampf, Marguerite C. Murphy, Rin Nakamura, Vilaiwan M. Fernandes, Darren E. Hagen, Shu Tao, Dan Graur, Florian Wolschin, Kirk J. Grubbs, Lumi Viljakainen, Neil D. Tsutsui, Cameron R. Currie, Chris Smith, Jay W. Kim, Christopher D. Smith, Mark Yandell, Ehab Abouheif, Hugh M. Robertson, Christine G. Elsik, Brian R. Johnson, Julie A. Mustard, Oliver Niehuis, Justin T. Reese, Carson Holt, Joshua D. Gibson, Elizabeth Cash, Kaitlyn A. Mathis, Marie-Julie Favé, Rick P. Overson, Rajendhran Rajakumar, Garret Suen, Wulfila Gronenberg, Aleksey V. Zimin, Candice W. Torres, Ana Sofia Ibarraran Viniegra, Eran Elhaik, Hao Hu, and Jennifer E. Placek
- Subjects
Whole genome sequencing ,Nasonia vitripennis ,Genetics ,Multidisciplinary ,biology ,Pseudogene ,Gene family ,Genomics ,Pogonomyrmex ,Red harvester ant ,Biological Sciences ,biology.organism_classification ,Genome - Abstract
We report the draft genome sequence of the red harvester ant, Pogonomyrmex barbatus . The genome was sequenced using 454 pyrosequencing, and the current assembly and annotation were completed in less than 1 y. Analyses of conserved gene groups (more than 1,200 manually annotated genes to date) suggest a high-quality assembly and annotation comparable to recently sequenced insect genomes using Sanger sequencing. The red harvester ant is a model for studying reproductive division of labor, phenotypic plasticity, and sociogenomics. Although the genome of P. barbatus is similar to other sequenced hymenopterans ( Apis mellifera and Nasonia vitripennis ) in GC content and compositional organization, and possesses a complete CpG methylation toolkit, its predicted genomic CpG content differs markedly from the other hymenopterans. Gene networks involved in generating key differences between the queen and worker castes (e.g., wings and ovaries) show signatures of increased methylation and suggest that ants and bees may have independently co-opted the same gene regulatory mechanisms for reproductive division of labor. Gene family expansions (e.g., 344 functional odorant receptors) and pseudogene accumulation in chemoreception and P450 genes compared with A. mellifera and N. vitripennis are consistent with major life-history changes during the adaptive radiation of Pogonomyrmex spp., perhaps in parallel with the development of the North American deserts.
- Published
- 2011
12. Abstract 4901: Comparison of gene expression platforms: RNA-Seq, Fluidigm, and Nanostring
- Author
-
Thomas Sandmann, Rin Nakamura, Rajesh Patel, Erica B. Schleifman, Craig Cummings, Ian McCaffery, An Do, Linda Bosch, Maipelo Motlhabi, Eric Peters, Andrew Watson, Walter C. Darbonne, and Rajiv Raja
- Subjects
Transcriptome ,Cancer Research ,Oncology ,Mrna level ,Gene expression ,RNA ,RNA-Seq ,Computational biology ,Biology ,DNA microarray ,Gene ,Molecular biology ,Biomarker (cell) - Abstract
Accurately measuring the expression of genes in formalin-fixed paraffin embedded (FFPE) tumor tissues has long been a struggle due to the inherent degradation of RNA isolated from these materials. Accurate quantification of gene expression levels in FFPE samples can enable the testing of biomarker hypotheses in the clinic and can potentially be used for patient stratification or selection in clinical trials. Platforms such as Fluidigm, Nanostring and microarrays are currently the high-throughput technologies utilized. Each of these platforms has different drawbacks and challenges such as lack of sensitivity, reproducibility or dynamic range when working with degraded FFPE samples. Currently, whole transcriptome RNA sequencing (RNA-Seq) is the only platform that offers a truly high-throughput, sensitive, and reproducible method to accurately quantify mRNA levels in RNA derived from FFPE samples. RNA-Seq, however has not yet been widely evaluated and adapted for use with degraded FFPE samples. Here we report the direct comparison of whole transcriptome RNA-Seq with two platforms that are currently compatible with FFPE derived RNA, Fluidigm and Nanostring, to determine the accuracy and feasibility of using this technology on degraded RNA. Utilizing a collection of matched fresh frozen (FF) and FFPE samples we analyzed gene expression using all three platforms to allow for a direct evaluation of each technology. By comparing the results to the matching FF sample, we were able to determine the accuracy and sensitivity of each platform when using degraded FFPE derived RNA. By titrating down the input of FFPE RNA into the RNA-Seq library prep we were also able to define the optimal input of FFPE RNA needed to accurately and reproducibly quantify gene expression. This work allows for a systematic comparison of different gene expression platforms for their use with degraded FFPE RNA. Citation Format: Erica B. Schleifman, Maipelo Motlhabi, Craig Cummings, Rin Nakamura, Linda Bosch, Rajesh Patel, An Do, Andrew Watson, Thomas Sandmann, Walter Darbonne, Ian McCaffery, Eric Peters, Rajiv Raja. Comparison of gene expression platforms: RNA-Seq, Fluidigm, and Nanostring. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 4901. doi:10.1158/1538-7445.AM2015-4901
- Published
- 2015
- Full Text
- View/download PDF
13. Circulating and tumor-based biomarkers predict clinical activity in cancer patients treated with the engineered anti-PD-L1 antibody MPDL3280A
- Author
-
Xiaodong Shen, Daniel S. Chen, Gregg Fine, Marcin Kowanetz, Rin Nakamura, Rajesh Patel, Marigold Boe, Rupal Desai, Jean-Charles Soria, Cecilia Leddy, Roy S. Herbst, Jane Ruppel, Priti S. Hegde, Hartmut Koeppen, Mitchell Denker, John D. Powderly, Ahmad Mokatrin, Yuanyuan Xiao, Qun J Wu, Ling Fu, Christina Rabe, Holbrook E Kohrt, Teiko Sumiyoshi, and Scott N. Gettinger
- Subjects
Pharmacology ,Cancer Research ,Tumor microenvironment ,biology ,business.industry ,Melanoma ,T cell ,Immunology ,Gene signature ,medicine.disease ,Immune checkpoint ,medicine.anatomical_structure ,Immune system ,Oncology ,Granzyme ,Poster Presentation ,biology.protein ,Molecular Medicine ,Immunology and Allergy ,Medicine ,Antibody ,business - Abstract
PD-L1 expressed in the tumor microenvironment regulates Th1 immune responses and mediates cancer immune evasion through interactions with PD-1 or B7.1 receptors on activated T cells. MPDL3280A, an engineered human monoclonal antibody, targets PD-L1 and inhibits its function. To identify immunologic predictive and pharmacodynamic biomarkers of MPDL3280A treatment, we performed a comprehensive analysis of tumors and blood samples collected at baseline and/or on treatment from ≈280 patients with locally advanced or metastatic solid tumors, including NSCLC, RCC, melanoma and bladder cancer. Regardless of tumor type, clinical responses were characterized by PD-L1 expression, the presence of markers of T cell activation (Th1 gene signature and CTLA4), and the absence of fractalkine at baseline in the tumor microenvironment. Elevated baseline expression of IFN-γ and IFN-γ-inducible genes (e.g., IDO1 and CXCL9) was associated with MPDL3280A response in melanoma but not NSCLC or RCC. On treatment, responding tumors showed increased infiltration of Th1-dominant immune infiltrate and evidence of adaptive PD-L1 up-regulation. In contrast, progressing tumors displayed the following patterns of tumor-infiltrating lymphocytes (TILs) and PD-L1 expression: (1) few/no TILs and absent PD-L1 expression (immunologic ignorance), (2) TILs present with minimal/no PD-L1 expression (non-functional immune responses), or (3) TILs residing solely around the tumor cell mass outer edge (excluded infiltrate), suggesting that resistance to MPDL3280A may be associated with impaired T cell trafficking and/or function. Profiling of ≈180 circulating biomarkers revealed that plasma concentrations of IL-18 and interferon-inducible T cell alpha chemoattractant (ITAC) increased in all patients following MPDL3280A treatment, representing a pharmacodynamic measurement of PD-L1 inhibition. In addition, analysis of PBMC showed an increase in T cell activation, as measured by IFN-γ and granzymes A and B gene expression in responders following MPDL3280A treatment, consistent with the observations in responding tumors. Baseline soluble PD-L1 was not associated with response. Some indication-specific biomarkers, such as plasma VEGF, decreased in responders with RCC but not with other indications. In NSCLC, a decrease in tumor burden markers, CA-125 and CEA, was associated with response. Similarly, IL-6 and IL-8 were differentially expressed on treatment in responders vs non-responders. Additionally, responders exhibited a decrease in circulating tumor DNA (ctDNA) in plasma, suggesting that ctDNA may be used to monitor MPDL3280A clinical activity in NSCLC. In conclusion, these data provide general and indication-specific mechanistic insights into immune checkpoint inhibition, potential mechanisms of response and resistance, as well as identification of potential predictive and pharmacodynamic biomarkers of anti-PD-L1/PD-1 clinical activity across multiple tumor types.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.