66 results on '"Robin Parihar"'
Search Results
2. Selectively targeting myeloid-derived suppressor cells through TRAIL receptor 2 to enhance the efficacy of CAR T cell therapy for treatment of breast cancer
- Author
-
Robin Parihar, Stephen Gottschalk, Ann Leen, Pradip Bajgain, Norihiro Watanabe, Valentina Hoyos, Mary K McKenna, Lauren Kelly, Paul Shafer, Saisha A Nalawade, Arushana Ali, Jarrett Joubert, and Juan Fernando Vera Valdes
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2021
- Full Text
- View/download PDF
3. Book of Abstracts: 2019 Health Equity Summer Research Summit Organized by the Center of Excellence in Health Equity, Training and Research, Baylor College of Medicine, Houston, Texas 77030, USA on June 18th, 2019
- Author
-
A Uribe-Gomez, Abraham A. Salinas-Miranda, MD, PhD, Acara E. Turner, BS, Adriana M. Strutt, PhD, ABPP, Adrienne Joseph, BS, Alex Wright, BS, Alexander C. Martin, BS, Alexis N. Milton, BS, Amelia Khoei, BS, Amritha Kanakamedala, BA, Angie Iyinbor, BA, Anna O’Neal, BS, Asha Morrow, MD, Ashley M. Butler, PhD, Ashley Young, BS, Asim Shah, MD, Asna Matin, MD, Audrey E. Christiansen, PhD, Boubakari Ibrahimou, PhD, Breanna Alonzo, Chase Ossenkop, BS, Carli O’Neal, BS, Chih-Wei Hsu, PhD, Charles Ikeanyionwu, BS, Chinwe Anyanwu, MPH, Charlotte Rivas, BS, Chioma A. Ikedionwu, BS, Chioma Anugwom, BS, Christopher R. Broda, MD, Claire Bocchini, MD, Claire Cummins, MD, Cliona M. Rooney, PhD, Colton L. Keo-Meier, PhD, Connor Johnson, BS, Courtney Miller-Chism, MD, Courtney Titus, MEd, Crystal L. Parker, MPH, Cylaina Bird, BA, Daniel Mauck, MPH, David Persse, MD, FACEP, Deborah A. Austin, PhD, Deepa Dongarwar, MS, Deepa V. Cherla, MD, Dwayne Wolf, MD, Ph.D, Eleanor Bimla Schwartz, MD, MS, Elena Petrova, PhD, Elias Perli, BS, Elisabeth Shell, PhD, PA-C, Elizabeth U. Tran, MD, Elsa Baena, PhD, Elyse Lopez, BS, Estrellita “Lo” Berry, MPA, Evadne Rodriguez, BA, Fabrizia Faustinella, MD, PhD, FACP, Faith D. Ihekweazu, MD, MS, Faiz Jiwani, MD, MS, Gabriella P. Chmaitelli, BS, Galant Chan, MD, Haijun Wang, PhD, MPH, Hamisu M. Salihu, MD, PhD, Hannah L. Combs, PhD, Hayley Rogers, BS, Heather Haq, MD, MHS, Iberia Romina Sosa, MD, PhD, Irene E. Jose, BS, Isabel Griffin, MPH, J. Cao, Jane Montealegre, PhD, Jason L. Salemi, PhD, MPH, Jennifer Chang, PhD, Jennifer M. Stinson, PhD, JL Mills, Joann Schulte, DO, MPH, Joel Thomas, John Prochaska, PhD, John Saunders, MD, MS, Jonathan Go, BBA, Jonathan Lim, MD, Jordan Salley, BS, Josue Estiven Pineda, BS, Jules Tabilona, MD, Julie L. Holihan, MD, MS, Kanika Bowen-Jallow, MD, MMS, Karla Bernardi, MD, Karla Fredricks, MD, MPH, Katherine B. Salciccioli, MD, Keila N. Lopez, MD,MPH, Kenneth Barning, MD, Kevin Dat Vuong, MA, Korede K. Yusuf, MBBS, PhD, Kristopher Myers, MPH, Kyle Wilson, Larry E. Laufman, EdD, Latanya J. Love, MD, Lauren Nolan, BA, Lauren Schoen, Lillian Huang, MD, Lindsey M. King, PhD, MPH, Lisa F. Nunez, BS, Louis Brown, PhD, Luna Hernandez, Lynn Hydod, M. Agustina Rossetti, PhD, Madeleine Allman, MPH, Mahmood Khan, MD, Makenna Marty, MAS, MaKenzie D. Lee, BS, Maria A. Jaramillo, BS, Maria Vigil, BS, Mariaelena Boyle, Marina Masciale, MD, Marisa Hilliard, PhD, Marisela Munoz, Mary E. Dickinson, PhD, Meishon Bell, MS, Michele K. York, PhD, Michelle Loor, MD, Michelle Lopez, MD, MPH, Mike K. Liang, MD, Moez Karim Aziz, MSECE, Moriel Karla, BS, Nancy Osazuma, Natalya Ramirez, BS, Natasha Navejar, BS, Nicolás Cortés-Penfield, MD, Nicole B. Lyons, BS, Norma Perez, MD, DrPH, NR Barshes, Olivia A. Barron, BA, Oscar A. Olavarria, MD, Petra Constable, BA, Phyllis Nwokolo, BS, Precious Omokaro, BS, Premal Patel, MD, Prithvi Vallabh, BS, Puja Shah, BA, Rafeek A. Yusuf, MD, MS, MPH, Rebecca A. Rosero, BA, Rebecca Lunstroth, JD, MA, Renice Obure, MPH, Robert Beach, MD, Robin Goin-Kochel, PhD, Robin Parihar, MD, PhD, Roger Zoorob, MD, MPH, Ronee E. Wilson, PhD, Rosa Michelle Schmidt, MD, MPH, Rowland Pettit, BS, Sade C. Udoetuk, MD, Sadia Tasnim, Sara Khan, BA, Sareema Adnan, BS, Sean Rodriguez, BA, Sharmila Anandasabapathy, MD, Shawna Nesbitt, MD, Sheena Bhushan, MD, Sherene Sharath, SO Rogers, Son Dinh, BSA, Sophia Banu, MD, Stacey Rose, MD, FACP, Stacy Drake, Ph.D, MPH, RN, AFN-BC, D-AB, Stephanie Morain, PhD, MPH, Stephen R. McCauley, PhD, Sue Abdelaziz, BS, Tahir Malik, BS, Tara L. Rasmussen, PhD, Thomas Shum, Tiana Raphel, BA, Tien C. Ko, MD, Toi Harris, MD, Trevor Hadley, BS, Uchechukwu Akoma, BS, Uwem Bridgette Eduok, BSA, Victoria Armendariz, MS, Yasmin C. Cole-Lewis, PhD, MPH, Zaina Al-Mohtaseb, MD, and Zenab Yusuf, MD, MPH
- Subjects
Public aspects of medicine ,RA1-1270 - Abstract
Copyright © 2020 Harris. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
- Published
- 2020
- Full Text
- View/download PDF
4. Chimeric Antigen Receptor Signaling Domains Differentially Regulate Proliferation and Native T Cell Receptor Function in Virus-Specific T Cells
- Author
-
Bilal Omer, Paul A. Castillo, Haruko Tashiro, Thomas Shum, Mai T. A. Huynh, Mara Cardenas, Miyuki Tanaka, Andrew Lewis, Tim Sauer, Robin Parihar, Natalia Lapteva, Michael Schmueck-Henneresse, Malini Mukherjee, Stephen Gottschalk, and Cliona M. Rooney
- Subjects
adoptive cell therapy ,chimeric antigen receptor ,virus specific t cells ,t cell receptor ,signaling domains ,Medicine (General) ,R5-920 - Abstract
The efficacy of T cells expressing chimeric antigen receptors (CARs) for solid tumors has been limited by insufficient CAR T cell expansion and persistence. The use of virus-specific T cells (VSTs) as carriers for CARs may overcome this limitation since CAR-VSTs can be boosted by viral vaccines or oncolytic viruses. However, there is limited understanding of the optimal combination of endodomains and their influence on the native T cell receptor (TCR) in VSTs. We therefore compared the function of GD2.CARs expressing the TCR zeta chain (ζ) alone or combined with endodomains from CD28 and 4-1BB in varicella zoster virus-specific (VZV) T cells. VZVSTs expressing GD2-CARs recognized VZV-derived peptides and killed GD2-expressing tumor cells. However, after repeated stimulation through their native TCR, the expansion of GD2-CAR.CD28ζ-VZVSTs was 3.3-fold greater (p < 0.001) than non-transduced VZVSTs, whereas GD2-CARζ- and GD2-CAR.41BBζ inhibited VZVST expansion (p < 0.01). Compared to control VZVSTs, GD2-CAR.ζ VZVSTs showed a greater frequency of apoptotic (p < 0.01) T cells, whereas prolonged downregulation of the native αβ TCR was observed in GD2-CAR.41BBζ VZVSTs (p < 0.001). We confirmed that CD28ζ can best maintain TCR function by expressing GD2.CARs in Epstein-Barr virus-specific T cells and CD19-CARs in VZVSTs. In response to CAR stimulation VSTs with CD28ζ endodomains also showed the greatest expansion (6 fold > GD2-CAR.41BBζ VZVSTs (p < 0.001), however anti-tumor efficacy was superior in GD2-CAR.41BBζ-VZVSTs. These findings demonstrate that CAR signaling domains can enhance or diminish the function of the native TCR and indicate that only CD28ζ may preserve the function of the native TCR in tonically signaling CAR-VSTs.
- Published
- 2018
- Full Text
- View/download PDF
5. TGF-β1 programmed myeloid-derived suppressor cells (MDSC) acquire immune-stimulating and tumor killing activity capable of rejecting established tumors in combination with radiotherapy
- Author
-
Padmini Jayaraman, Falguni Parikh, Jared M. Newton, Aurelie Hanoteau, Charlotte Rivas, Rosemarie Krupar, Kimal Rajapakshe, Ravi Pathak, Kavin Kanthaswamy, Cassie MacLaren, Shixia Huang, Cristian Coarfa, Chad Spanos, Dean P. Edwards, Robin Parihar, and Andrew G. Sikora
- Subjects
mdsc ,tgf-beta ,cd86 ,tumor killing ,radiotherapy ,caspase-3 ,myeloid-derived suppressor cells (written out) ,smad2 ,adoptive cellular therapy ,Immunologic diseases. Allergy ,RC581-607 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Cancer-induced myeloid-derived suppressor cells (MDSC) play an important role in tumor immune evasion. MDSC programming or polarization has been proposed as a strategy for leveraging the developmental plasticity of myeloid cells to reverse MDSC immune suppressive functions, or cause them to acquire anti-tumor activity. While MDSC derived ex vivo from murine bone marrow precursor cells with tumor-conditioned medium efficiently suppressed T cell proliferation, MDSC derived from conditioned medium in presence of TGF-β1 (TGFβ-MDSC) acquired a novel immune-stimulatory phenotype, losing the ability to inhibit T cell proliferation and acquiring enhanced antigen-presenting capability. Altered immune function was associated with SMAD-2 dependent upregulation of maturation and costimulatory molecules, and downregulation of inducible nitric oxide synthase (iNOS), an effector mechanism of immunosuppression. TGFβ-MDSC also upregulated FAS-ligand expression, leading to FAS-dependent killing of murine human papillomavirus (HPV)-associated head and neck cancer cells and tumor spheroids in vitro and anti-tumor activity in vivo. Radiation upregulated FAS expression on tumor cells, and the combination of radiotherapy and intratumoral injection of TGFβ-MDSC strongly enhanced class I expression on tumor cells and induction of HPV E7 tetramer-positive CD8 + T cells, leading to clearance of established tumors and long-term survival. TGFβ-MDSC derived from human PBMC with tumor conditioned medium also lost immunosuppressive function and acquired tumor-killing activity. Thus, TGFβ1 mediated programming of nascent MDSC leads to a potent anti-tumor phenotype potentially suitable for adoptive immunotherapy.
- Published
- 2018
- Full Text
- View/download PDF
6. Supplementary Data from NK Cells Expressing a Chimeric Activating Receptor Eliminate MDSCs and Rescue Impaired CAR-T Cell Activity against Solid Tumors
- Author
-
Cliona M. Rooney, Stephen M. Gottschalk, Leonid S. Metelitsa, Natalia Lapteva, Bilal Omer, Mai Huynh, Charlotte Rivas, and Robin Parihar
- Abstract
Fig. S1 shows characterization of GD2-specific CAR-T cells. Fig. S2 shows characterization of human MDSCs. Fig. S3 shows characterization of TME xenograft model. Fig. S4 shows GD2.CAR-T cell homing and infiltration into MDSC-containing tumors after NK cell infusion. Fig. S5 shows infiltration of NK cell and GD2.CAR-T cells by IHC.
- Published
- 2023
- Full Text
- View/download PDF
7. Data from NK Cells Expressing a Chimeric Activating Receptor Eliminate MDSCs and Rescue Impaired CAR-T Cell Activity against Solid Tumors
- Author
-
Cliona M. Rooney, Stephen M. Gottschalk, Leonid S. Metelitsa, Natalia Lapteva, Bilal Omer, Mai Huynh, Charlotte Rivas, and Robin Parihar
- Abstract
Solid tumors are refractory to cellular immunotherapies in part because they contain suppressive immune effectors such as myeloid-derived suppressor cells (MDSCs) that inhibit cytotoxic lymphocytes. Strategies to reverse the suppressive tumor microenvironment (TME) should also attract and activate immune effectors with antitumor activity. To address this need, we developed gene-modified natural killer (NK) cells bearing a chimeric receptor in which the activating receptor NKG2D is fused to the cytotoxic ζ-chain of the T-cell receptor (NKG2D.ζ). NKG2D.ζ–NK cells target MDSCs, which overexpress NKG2D ligands within the TME. We examined the ability of NKG2D.ζ–NK cells to eliminate MDSCs in a xenograft TME model and improve the antitumor function of tumor-directed chimeric antigen receptor (CAR)–modified T cells. We show that NKG2D.ζ–NK cells are cytotoxic against MDSCs, but spare NKG2D ligand–expressing normal tissues. NKG2D.ζ–NK cells, but not unmodified NK cells, secrete proinflammatory cytokines and chemokines in response to MDSCs at the tumor site and improve infiltration and antitumor activity of subsequently infused CAR-T cells, even in tumors for which an immunosuppressive TME is an impediment to treatment. Unlike endogenous NKG2D, NKG2D.ζ is not susceptible to TME-mediated downmodulation and thus maintains its function even within suppressive microenvironments. As clinical confirmation, NKG2D.ζ–NK cells generated from patients with neuroblastoma killed autologous intratumoral MDSCs capable of suppressing CAR-T function. A combination therapy for solid tumors that includes both NKG2D.ζ–NK cells and CAR-T cells may improve responses over therapies based on CAR-T cells alone.
- Published
- 2023
- Full Text
- View/download PDF
8. Supplemental Figure 1 from Constitutive Signaling from an Engineered IL7 Receptor Promotes Durable Tumor Elimination by Tumor-Redirected T Cells
- Author
-
Cliona M. Rooney, Stephen Gottschalk, Leonid S. Metelitsa, Malcolm K. Brenner, Hao Liu, Paul Castillo, Robin Parihar, Daofeng Liu, Tim Sauer, Zhongzhen Yi, Kathan Parikh, Dimitrios L. Wagner, Robert L. Kruse, Haruko Tashiro, Bilal Omer, and Thomas Shum
- Abstract
Supplementary Figure 1: CD34-IL7R* (C7R) is stably expressed in T-cells and enhances constitutive STAT5 activation relative to IL7R*.
- Published
- 2023
- Full Text
- View/download PDF
9. Supplemental Figure 3 from Constitutive Signaling from an Engineered IL7 Receptor Promotes Durable Tumor Elimination by Tumor-Redirected T Cells
- Author
-
Cliona M. Rooney, Stephen Gottschalk, Leonid S. Metelitsa, Malcolm K. Brenner, Hao Liu, Paul Castillo, Robin Parihar, Daofeng Liu, Tim Sauer, Zhongzhen Yi, Kathan Parikh, Dimitrios L. Wagner, Robert L. Kruse, Haruko Tashiro, Bilal Omer, and Thomas Shum
- Abstract
Supplementary Figure 3: C7R signaling does not change phenotypic composition of GD2-CAR T-cells during culture.
- Published
- 2023
- Full Text
- View/download PDF
10. Supplemental Figure 6 from Constitutive Signaling from an Engineered IL7 Receptor Promotes Durable Tumor Elimination by Tumor-Redirected T Cells
- Author
-
Cliona M. Rooney, Stephen Gottschalk, Leonid S. Metelitsa, Malcolm K. Brenner, Hao Liu, Paul Castillo, Robin Parihar, Daofeng Liu, Tim Sauer, Zhongzhen Yi, Kathan Parikh, Dimitrios L. Wagner, Robert L. Kruse, Haruko Tashiro, Bilal Omer, and Thomas Shum
- Abstract
Supplementary Figure 6: Efficient retroviral transduction of T-cells with Î"34, C7R, GD2-CAR, and GD2-CAR.C7R constructs.
- Published
- 2023
- Full Text
- View/download PDF
11. Supplemental Figure 5 from Constitutive Signaling from an Engineered IL7 Receptor Promotes Durable Tumor Elimination by Tumor-Redirected T Cells
- Author
-
Cliona M. Rooney, Stephen Gottschalk, Leonid S. Metelitsa, Malcolm K. Brenner, Hao Liu, Paul Castillo, Robin Parihar, Daofeng Liu, Tim Sauer, Zhongzhen Yi, Kathan Parikh, Dimitrios L. Wagner, Robert L. Kruse, Haruko Tashiro, Bilal Omer, and Thomas Shum
- Abstract
Supplementary Figure 5: C7R does not significantly increase intracranial EphA2-CAR T-cell expansion against U373 tumors
- Published
- 2023
- Full Text
- View/download PDF
12. Supplemental Figures S1-5 from MICA-Expressing Monocytes Enhance Natural Killer Cell Fc Receptor-Mediated Antitumor Functions
- Author
-
William E. Carson, Michael A. Caligiuri, John C. Byrd, Vedat Yildiz, Lianbo Yu, Veronika Groh, Susheela Tridandapani, David Raulet, Robert Lee, Bryant Yung, Adrian Lewis, Eric Luedke, Robin Parihar, Prashant Trikha, Elizabeth L. McMichael, Kallan S. Opheim, Neela Bhave, Lorena P. Suarez-Kelly, Megan C. Duggan, and Amanda R. Campbell
- Abstract
Supplemental Figure S1. Contribution of NK and T cells to IFN-gamma production. Supplemental Figure S2. MICA expression on monocytes. Supplemental Figure S3. NKG2D Ligand Expression on C1R and C1R-MICA Cell Lines. Supplemental Figure S4. Monocyte MICA expression does not correlate with NK cell IFN-gamma production. Supplemental Figure S5. NKG2D and MICA expression levels prior to and following co-culture.
- Published
- 2023
- Full Text
- View/download PDF
13. Supplemental Figure 4 from Constitutive Signaling from an Engineered IL7 Receptor Promotes Durable Tumor Elimination by Tumor-Redirected T Cells
- Author
-
Cliona M. Rooney, Stephen Gottschalk, Leonid S. Metelitsa, Malcolm K. Brenner, Hao Liu, Paul Castillo, Robin Parihar, Daofeng Liu, Tim Sauer, Zhongzhen Yi, Kathan Parikh, Dimitrios L. Wagner, Robert L. Kruse, Haruko Tashiro, Bilal Omer, and Thomas Shum
- Abstract
Supplementary Figure 4: C7R prolongs survival of GD2-CAR T-cells but does not support autonomous cell expansion.
- Published
- 2023
- Full Text
- View/download PDF
14. Data from MICA-Expressing Monocytes Enhance Natural Killer Cell Fc Receptor-Mediated Antitumor Functions
- Author
-
William E. Carson, Michael A. Caligiuri, John C. Byrd, Vedat Yildiz, Lianbo Yu, Veronika Groh, Susheela Tridandapani, David Raulet, Robert Lee, Bryant Yung, Adrian Lewis, Eric Luedke, Robin Parihar, Prashant Trikha, Elizabeth L. McMichael, Kallan S. Opheim, Neela Bhave, Lorena P. Suarez-Kelly, Megan C. Duggan, and Amanda R. Campbell
- Abstract
Natural killer (NK) cells are large granular lymphocytes that promote the antitumor response via communication with other cell types in the tumor microenvironment. Previously, we have shown that NK cells secrete a profile of immune stimulatory factors (e.g., IFNγ, MIP-1α, and TNFα) in response to dual stimulation with the combination of antibody (Ab)-coated tumor cells and cytokines, such as IL12. We now demonstrate that this response is enhanced in the presence of autologous monocytes. Monocyte enhancement of NK cell activity was dependent on cell-to-cell contact as determined by a Transwell assay. It was hypothesized that NK cell effector functions against Ab-coated tumor cells were enhanced via binding of MICA on monocytes to NK cell NKG2D receptors. Strategies to block MICA–NKG2D interactions resulted in reductions in IFNγ production. Depletion of monocytes in vivo resulted in decreased IFNγ production by murine NK cells upon exposure to Ab-coated tumor cells. In mice receiving trastuzumab and IL12 therapy, monocyte depletion resulted in significantly greater tumor growth in comparison to mock-depleted controls (P < 0.05). These data suggest that NK cell–monocyte interactions enhance NK cell antitumor activity in the setting of monoclonal Ab therapy for cancer. Cancer Immunol Res; 5(9); 778–89. ©2017 AACR.
- Published
- 2023
- Full Text
- View/download PDF
15. Supplemental Table 1 from Constitutive Signaling from an Engineered IL7 Receptor Promotes Durable Tumor Elimination by Tumor-Redirected T Cells
- Author
-
Cliona M. Rooney, Stephen Gottschalk, Leonid S. Metelitsa, Malcolm K. Brenner, Hao Liu, Paul Castillo, Robin Parihar, Daofeng Liu, Tim Sauer, Zhongzhen Yi, Kathan Parikh, Dimitrios L. Wagner, Robert L. Kruse, Haruko Tashiro, Bilal Omer, and Thomas Shum
- Abstract
Supplemental Table 1: Differential gene expression in GD2-CAR.C7R T-cells compared to GD2-CAR T-cells.
- Published
- 2023
- Full Text
- View/download PDF
16. Supplemental Figure 2 from Constitutive Signaling from an Engineered IL7 Receptor Promotes Durable Tumor Elimination by Tumor-Redirected T Cells
- Author
-
Cliona M. Rooney, Stephen Gottschalk, Leonid S. Metelitsa, Malcolm K. Brenner, Hao Liu, Paul Castillo, Robin Parihar, Daofeng Liu, Tim Sauer, Zhongzhen Yi, Kathan Parikh, Dimitrios L. Wagner, Robert L. Kruse, Haruko Tashiro, Bilal Omer, and Thomas Shum
- Abstract
Supplementary Figure 2: Efficient retroviral transduction of C7R and Î"34 in both CD4 and CD8 selected T-cells.
- Published
- 2023
- Full Text
- View/download PDF
17. Contributors
- Author
-
Maria M. Abreu, Prasad S. Adusumilli, Behnam Badie, Rafet Basar, Rebecca Bellis, Ronald Blasberg, Barbara Breznik, Shyambabu Chaurasiya, Po-Chun Chen, Kevin Choong, Nishant Chovatiya, May Daher, Supriya Deshpande, Lisa Feldman, Timothy D. Folsom, Christina Fong, Yuman Fong, Jacqueline Gouvea, Anamarija Habič, Anahid Jewett, Sharyn I. Katz, Kawaljit Kaur, Meng-Wei Ko, Janko Kos, Derek Lee, Zhe Li, Emil Lou, Dipnarine Maharaj, Kelly Mahuron, Bernarda Majc, Sandro Matosevic, Branden S. Moriarity, John P. Murad, Ishwar Navin, Metka Novak, Robin Parihar, Anthony K. Park, Vineet Polineni, Vladimir Ponomarev, Saul J. Priceman, Hind Rafei, Jamie Rand, Katayoun Rezvani, Isabelle Rivière, Leonid Roshkovan, Saeed Sadeghi, Jasmeen Saini, Emanuela Senjor, Inna Serganova, Timothy K. Starr, Tamara Lah Turnsek, Alessia Volpe, Xiuyan Wang, Beau R. Webber, Paul Wong, Yanghee Woo, Jennifer Wu, Lili Yang, Yuan Yuan, Samuel Zeng, and Amy Zhu
- Published
- 2023
- Full Text
- View/download PDF
18. Reversing the NK inhibitory tumor microenvironment by targeting suppressive immune effectors
- Author
-
Ishwar Navin and Robin Parihar
- Published
- 2023
- Full Text
- View/download PDF
19. 328 Promoting NK survival and function within the tumor microenvironment
- Author
-
Matthew Dysthe, Ishwar Navin, Corrine Baumgartner, Stephanie Fetzko, Cliona Rooney, and Robin Parihar
- Published
- 2022
- Full Text
- View/download PDF
20. 816 Blockade of SIRPα on macrophages to enhance CAR T activity against glioblastoma
- Author
-
Rebecca Brock, Alesandra Echeandia-Marrero, Matthew Dysthe, Robin Parihar, Nabil Ahmed, and Meenakshi Hegde
- Published
- 2022
- Full Text
- View/download PDF
21. 413 Genetic deletion of TIGIT enhances CAR-NK cell function in the solid tumor microenvironment
- Author
-
Ishwar Navin, Matthew Dysthe, Corrine Baumgartner, and Robin Parihar
- Published
- 2022
- Full Text
- View/download PDF
22. 394 Using co-stimulatory CARs in natural killer cells to safely target soft-tissue sarcomas and their inhibitory microenvironments
- Author
-
Stephanie Fetzko, Ishwar Navin, Matthew Dysthe, Tim Sauer, Cliona Rooney, and Robin Parihar
- Published
- 2022
- Full Text
- View/download PDF
23. Oncolytic adeno-immunotherapy modulates the immune system enabling CAR T-cells to cure pancreatic tumors
- Author
-
Matthew Dysthe, Malcolm K. Brenner, Youngrock Jung, Tiffany Yip, Masataka Suzuki, Caroline E. Porter, Way-Champ Mah, Robin Parihar, Mary K. McKenna, and Amanda Rosewell Shaw
- Subjects
0301 basic medicine ,Male ,endocrine system diseases ,Receptor, ErbB-2 ,medicine.medical_treatment ,T-Lymphocytes ,Medicine (miscellaneous) ,Cancer immunotherapy ,Immunotherapy, Adoptive ,B7-H1 Antigen ,0302 clinical medicine ,Tumor Microenvironment ,Neoplasm Metastasis ,Biology (General) ,skin and connective tissue diseases ,Oncolytic Virotherapy ,Receptors, Chimeric Antigen ,Primary tumor ,Interleukin-12 ,Tumor Burden ,Oncolytic Viruses ,030220 oncology & carcinogenesis ,Female ,General Agricultural and Biological Sciences ,Carcinoma, Pancreatic Ductal ,QH301-705.5 ,General Biochemistry, Genetics and Molecular Biology ,Article ,Adenoviridae ,03 medical and health sciences ,Pancreatic cancer ,Cell Line, Tumor ,medicine ,Humans ,Tumor microenvironment ,business.industry ,Immunotherapy ,medicine.disease ,Xenograft Model Antitumor Assays ,Immune checkpoint ,Coculture Techniques ,digestive system diseases ,Oncolytic virus ,Pancreatic Neoplasms ,030104 developmental biology ,Humanized mouse ,Cancer research ,business ,Single-Chain Antibodies - Abstract
High expression levels of human epidermal growth factor receptor 2 (HER2) have been associated with poor prognosis in patients with pancreatic adenocarcinoma (PDAC). However, HER2-targeting immunotherapies have been unsuccessful to date. Here we increase the breadth, potency, and duration of anti-PDAC HER2-specific CAR T-cell (HER2.CART) activity with an oncolytic adeno-immunotherapy that produces cytokine, immune checkpoint blockade, and a safety switch (CAdTrio). Combination treatment with CAdTrio and HER2.CARTs cured tumors in two PDAC xenograft models and produced durable tumor responses in humanized mice. Modifications to the tumor immune microenvironment contributed to the antitumor activity of our combination immunotherapy, as intratumoral CAdTrio treatment induced chemotaxis to enable HER2.CART migration to the tumor site. Using an advanced PDAC model in humanized mice, we found that local CAdTrio treatment of primary tumor stimulated systemic host immune responses that repolarized distant tumor microenvironments, improving HER2.CART anti-tumor activity. Overall, our data demonstrate that CAdTrio and HER2.CARTs provide complementary activities to eradicate metastatic PDAC and may represent a promising co-operative therapy for PDAC patients., Rosewell Shaw et al. show that a previously developed immunotherapy strategy, coupling oncolytic adenoviral immunotherapy with clinically tested HER2-specific CAR T-cells, is effective against pancreatic ductal adenocarcinoma (PDAC). This combination therapy produces a curative response in both PDAC xenografts and humanized mouse models.
- Published
- 2021
24. Memory NK cells to forget relapsed AML
- Author
-
Robin Parihar
- Subjects
Killer Cells, Natural ,Leukemia, Myeloid, Acute ,Immunobiology and Immunotherapy ,Immunology ,Humans ,Cell Biology ,Hematology ,Biochemistry - Abstract
Pediatric and young adult (YA) patients with acute myeloid leukemia (AML) who relapse after allogeneic hematopoietic cell transplantation (HCT) have an extremely poor prognosis. Standard salvage chemotherapy and donor lymphocyte infusions (DLIs) have little curative potential. Previous studies showed that natural killer (NK) cells can be stimulated ex vivo with interleukin-12 (IL-12), -15, and -18 to generate memory-like (ML) NK cells with enhanced antileukemia responses. We treated 9 pediatric/YA patients with post-HCT relapsed AML with donor ML NK cells in a phase 1 trial. Patients received fludarabine, cytarabine, and filgrastim followed 2 weeks later by infusion of donor lymphocytes and ML NK cells from the original HCT donor. ML NK cells were successfully generated from haploidentical and matched-related and -unrelated donors. After infusion, donor-derived ML NK cells expanded and maintained an ML multidimensional mass cytometry phenotype for >3 months. Furthermore, ML NK cells exhibited persistent functional responses as evidenced by leukemia-triggered interferon-γ production. After DLI and ML NK cell adoptive transfer, 4 of 8 evaluable patients achieved complete remission at day 28. Two patients maintained a durable remission for >3 months, with 1 patient in remission for >2 years. No significant toxicity was experienced. This study demonstrates that, in a compatible post-HCT immune environment, donor ML NK cells robustly expand and persist with potent antileukemic activity in the absence of exogenous cytokines. ML NK cells in combination with DLI present a novel immunotherapy platform for AML that has relapsed after allogeneic HCT. This trial was registered at https://clinicaltrials.gov as #NCT03068819.
- Published
- 2021
25. NK Cells Expressing a Chimeric Activating Receptor Eliminate MDSCs and Rescue Impaired CAR-T Cell Activity against Solid Tumors
- Author
-
Natalia Lapteva, Robin Parihar, Mai Huynh, Leonid S. Metelitsa, Charlotte H. Rivas, Cliona M. Rooney, Bilal Omer, and Stephen Gottschalk
- Subjects
Cytotoxicity, Immunologic ,0301 basic medicine ,Cancer Research ,Recombinant Fusion Proteins ,T-Lymphocytes ,medicine.medical_treatment ,Immunology ,Receptors, Antigen, T-Cell ,chemical and pharmacologic phenomena ,Ligands ,Immunotherapy, Adoptive ,Article ,Mice ,Neuroblastoma ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antigen ,Cell Line, Tumor ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Cytotoxic T cell ,Tumor microenvironment ,Receptors, Chimeric Antigen ,Chemistry ,Myeloid-Derived Suppressor Cells ,hemic and immune systems ,Immunotherapy ,NKG2D ,Xenograft Model Antitumor Assays ,biological factors ,Chimeric antigen receptor ,Killer Cells, Natural ,030104 developmental biology ,NK Cell Lectin-Like Receptor Subfamily K ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Chemokines ,K562 Cells ,K562 cells - Abstract
Solid tumors are refractory to cellular immunotherapies in part because they contain suppressive immune effectors such as myeloid-derived suppressor cells (MDSCs) that inhibit cytotoxic lymphocytes. Strategies to reverse the suppressive tumor microenvironment (TME) should also attract and activate immune effectors with antitumor activity. To address this need, we developed gene-modified natural killer (NK) cells bearing a chimeric receptor in which the activating receptor NKG2D is fused to the cytotoxic ζ-chain of the T-cell receptor (NKG2D.ζ). NKG2D.ζ–NK cells target MDSCs, which overexpress NKG2D ligands within the TME. We examined the ability of NKG2D.ζ–NK cells to eliminate MDSCs in a xenograft TME model and improve the antitumor function of tumor-directed chimeric antigen receptor (CAR)–modified T cells. We show that NKG2D.ζ–NK cells are cytotoxic against MDSCs, but spare NKG2D ligand–expressing normal tissues. NKG2D.ζ–NK cells, but not unmodified NK cells, secrete proinflammatory cytokines and chemokines in response to MDSCs at the tumor site and improve infiltration and antitumor activity of subsequently infused CAR-T cells, even in tumors for which an immunosuppressive TME is an impediment to treatment. Unlike endogenous NKG2D, NKG2D.ζ is not susceptible to TME-mediated downmodulation and thus maintains its function even within suppressive microenvironments. As clinical confirmation, NKG2D.ζ–NK cells generated from patients with neuroblastoma killed autologous intratumoral MDSCs capable of suppressing CAR-T function. A combination therapy for solid tumors that includes both NKG2D.ζ–NK cells and CAR-T cells may improve responses over therapies based on CAR-T cells alone.
- Published
- 2019
- Full Text
- View/download PDF
26. Detection of response to tumor microenvironment–targeted cellular immunotherapy using nano-radiomics
- Author
-
Ananth Annapragada, Robin Parihar, Ketan B. Ghaghada, Zbigniew Starosolski, Laxman Devkota, Charlotte H. Rivas, and Igor Stupin
- Subjects
medicine.medical_treatment ,Immunology ,Cell ,Tumor response ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Radiomics ,law ,Neoplasms ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Solid tumor ,Research Articles ,Cancer ,030304 developmental biology ,0303 health sciences ,Tumor microenvironment ,Multidisciplinary ,business.industry ,Myeloid-Derived Suppressor Cells ,SciAdv r-articles ,Immunotherapy ,Killer Cells, Natural ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer research ,Suppressor ,sense organs ,Cellular immunotherapy ,business ,Research Article - Abstract
Data mining of nanoparticle contrast–enhanced 3D images reveals phenotypes indicative of tumor response to cellular immunotherapy., Immunotherapies, including cell-based therapies, targeting the tumor microenvironment (TME) result in variable and delayed responses. Thus, it has been difficult to gauge the efficacy of TME-directed therapies early after administration. We investigated a nano-radiomics approach (quantitative analysis of nanoparticle contrast–enhanced three-dimensional images) for detection of tumor response to cellular immunotherapy directed against myeloid-derived suppressor cells (MDSCs), a key component of TME. Animals bearing human MDSC-containing solid tumor xenografts received treatment with MDSC-targeting human natural killer (NK) cells and underwent nanoparticle contrast–enhanced computed tomography (CT) imaging. Whereas conventional CT-derived tumor metrics were unable to differentiate NK cell immunotherapy tumors from untreated tumors, nano-radiomics revealed texture-based features capable of differentiating treatment groups. Our study shows that TME-directed cellular immunotherapy causes subtle changes not effectively gauged by conventional imaging metrics but revealed by nano-radiomics. Our work provides a method for noninvasive assessment of TME-directed immunotherapy potentially applicable to numerous solid tumors.
- Published
- 2020
- Full Text
- View/download PDF
27. Myeloid-Derived Suppressor Cells in the Tumor Microenvironment
- Author
-
Matthew, Dysthe and Robin, Parihar
- Subjects
Myeloid-Derived Suppressor Cells ,Neoplasms ,Tumor Microenvironment ,Humans ,Myeloid Cells ,Tumor Escape - Abstract
Myeloid-derived suppressor cells (MDSCs) represent a heterogenous population of immature myeloid cells capable of modulating immune responses. In the context of cancer, MDSCs are abnormally produced and recruited to the tumor microenvironment (TME) to aid in the establishment of an immunosuppressive TME that facilitates tumor escape. Additionally, MDSCs contribute to non-immunologic aspects of tumor biology, including tumor angiogenesis and metastasis. The clinical significance of MDSCs has recently been appreciated as numerous studies have suggested a correlation between circulating and intratumoral MDSC frequencies and tumor stage, progression, and treatment resistance. In this chapter, we review MDSC characterization, development, expansion, and mechanisms that facilitate immunosuppression and tumor progression. Furthermore, we highlight studies demonstrating the clinical significance of MDSCs in various disease states in addition to strategies that modulate various aspects of MDSC biology for therapeutic gain.
- Published
- 2020
28. Overcoming the breast tumor microenvironment by targeting MDSCs through CAR-T cell therapy
- Author
-
Robin Parihar, Juan Fernando Vera Valdes, Ann M. Leen, Katie McKenna, Lauren Kelly, Saisha Abhay Nalawade, Valentina Hoyos Velez, Arushana Ali, Pradip Bajgain, Jarrett Joubert, Paul Shafer, and Norihiro Watanabe
- Subjects
Cancer Research ,Tumor microenvironment ,Immune system ,Breast cancer ,Oncology ,business.industry ,Cancer research ,Medicine ,CAR T-cell therapy ,Car t cells ,business ,medicine.disease ,Breast tumor - Abstract
1032 Background: Successful targeting of solid tumors such as breast cancer (BC) using CAR T cells (CARTs) has proven challenging, largely due to the immune suppressive tumor microenvironment (TME). Myeloid derived suppressor cells (MDSCs) inhibit CART’s function and persistence within the breast TME. We generated CAR T cells targeting tumor-expressed mucin 1 (MUC1) (Bajgain P et al, 2018) for BC. To potentiate expansion and persistence of MUC1 CARTs and modulate the suppressive TME, we developed a novel chimeric co-stimulatory receptor, TR2.4-1BB, encoding a ScFv derived from a TNF-related apoptosis-inducing ligand receptor 2 (TR2) mAb followed by a 4-1BB endodomain. We hypothesize that engagement with TR2 expressed on TME-resident MDSCs, will lead to both MDSC apoptosis and CART co-stimulation, promoting T cell persistence and expansion at tumor site. Methods: Function of the novel TR2.4-1BB receptor, was assessed by exposing non-transduced (NT) and TR2.4-1BB transduced T cells to recombinant TR2 and nuclear translocation of NFκB was measured by ELISA. Functionality of in vitro generated MDSCs was determined by the suppression assay. In vitro CART/costimulatory receptor T cell function was measured by cytotoxicity assays using MUC1+ tumor targets in presence or absence of MDSCs. In vivo anti-tumor activity was assessed using MDSC enriched tumor-bearing mice using calipers to assess tumor volume and bioluminescence imaging to track T cells. Results: Nuclear translocation of NFκB was detected only in TR2.4-1BB T cells. MDSCs significantly attenuated T cell proliferation by 50±5% and IFNγ production by half compared with T cells cultured alone. Additionally, presence of MDSCs, diminished cytotoxic potential of MUC1 CARTs against MUC1+ BC cell lines by 25%. However, TR2.4-1BB expression on CAR.MUC1 T cells induced MDSC apoptosis thereby restoring the cytotoxic activity of CAR.MUC1 against MUC1+ BC lines in presence of TR2.4-1BB (67±8.5%). There was an approximate two-fold increase in tumor growth due enhanced angiogenesis and fibroblast accumulation in mice receiving tumors + MDSCs compared to tumors alone. Treatment of these MDSC-enriched tumors with MUC1.TR2.4-1BB CARTs led to superior tumor cell killing and significant reduction in tumor growth (24.54±8.55 mm3) compared to CAR.MUC1 (469.79.9±81.46mm3) or TR2.4-1BB (434.86±64.25 mm3) T cells alone (Day 28 after T cell injection). The treatment also improved T cell proliferation and persistence at the tumor site. Thereby, leading to negligible metastasis demonstrating ability of CARTs to eliminate tumor and prevent dissemination. We observed similar results using HER2.TR2.4-1BB CARTs in a HER2+ BC model. Conclusions: Our findings demonstrate that CARTs co-expressing our novel TR2.4-1BB receptor have higher anti-tumor potential against BC tumors and infiltrating MDSCs, resulting in TME remodeling and improved T cell proliferation at the tumor site.
- Published
- 2021
- Full Text
- View/download PDF
29. MICA-expressing monocytes enhance natural killer cell Fc receptor-mediated antitumor functions
- Author
-
Adrian Lewis, Prashant Trikha, Robin Parihar, Lianbo Yu, Vedat O. Yildiz, Amanda Campbell, Robert J. Lee, Neela Bhave, Megan C. Duggan, William E. Carson, David H. Raulet, Susheela Tridandapani, Bryant C. Yung, Kallan S. Opheim, Elizabeth L. McMichael, Lorena P. Suarez-Kelly, Veronika Groh, Michael A. Caligiuri, John C. Byrd, and Eric Luedke
- Subjects
0301 basic medicine ,Cancer Research ,Oncology and Carcinogenesis ,Immunology ,Breast Neoplasms ,Receptors, Fc ,Biology ,Article ,Monocytes ,Antibodies ,Cell Line ,Natural killer cell ,Vaccine Related ,Mice ,Interferon-gamma ,03 medical and health sciences ,Interleukin 21 ,Cell Line, Tumor ,Receptors ,Monoclonal ,medicine ,Killer Cells ,Animals ,Humans ,2.1 Biological and endogenous factors ,Aetiology ,Cancer ,Tumor ,Lymphokine-activated killer cell ,Fc ,Monocyte ,Histocompatibility Antigens Class I ,Autologous Monocytes ,Antibodies, Monoclonal ,Pharmacology and Pharmaceutical Sciences ,Trastuzumab ,NKG2D ,Interleukin-12 ,Killer Cells, Natural ,030104 developmental biology ,medicine.anatomical_structure ,NK Cell Lectin-Like Receptor Subfamily K ,Natural ,Cancer research ,Interleukin 12 ,Female ,Tumor necrosis factor alpha - Abstract
Natural killer (NK) cells are large granular lymphocytes that promote the antitumor response via communication with other cell types in the tumor microenvironment. Previously, we have shown that NK cells secrete a profile of immune stimulatory factors (e.g., IFNγ, MIP-1α, and TNFα) in response to dual stimulation with the combination of antibody (Ab)-coated tumor cells and cytokines, such as IL12. We now demonstrate that this response is enhanced in the presence of autologous monocytes. Monocyte enhancement of NK cell activity was dependent on cell-to-cell contact as determined by a Transwell assay. It was hypothesized that NK cell effector functions against Ab-coated tumor cells were enhanced via binding of MICA on monocytes to NK cell NKG2D receptors. Strategies to block MICA–NKG2D interactions resulted in reductions in IFNγ production. Depletion of monocytes in vivo resulted in decreased IFNγ production by murine NK cells upon exposure to Ab-coated tumor cells. In mice receiving trastuzumab and IL12 therapy, monocyte depletion resulted in significantly greater tumor growth in comparison to mock-depleted controls (P < 0.05). These data suggest that NK cell–monocyte interactions enhance NK cell antitumor activity in the setting of monoclonal Ab therapy for cancer. Cancer Immunol Res; 5(9); 778–89. ©2017 AACR.
- Published
- 2017
- Full Text
- View/download PDF
30. Constitutive signaling from an engineered IL-7 receptor promotes durable tumor elimination by tumor redirected T-cells
- Author
-
Dimitrios L. Wagner, Robin Parihar, Kathan Parikh, Zhongzhen Yi, Haruko Tashiro, Malcolm K. Brenner, Stephen Gottschalk, Paul Castillo, Thomas Shum, Robert L. Kruse, Leonid S. Metelitsa, Bilal Omer, Daofeng Liu, Hao Liu, Cliona M. Rooney, and Tim Sauer
- Subjects
0301 basic medicine ,medicine.medical_treatment ,T-Lymphocytes ,Cell ,Receptors, Antigen, T-Cell ,Biology ,Immunotherapy, Adoptive ,Article ,03 medical and health sciences ,Mice ,Neuroblastoma ,Antigen ,Antigens, Neoplasm ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Receptors, Cytokine ,Receptor ,Cell Proliferation ,Interleukin-7 ,Immunotherapy ,Xenograft Model Antitumor Assays ,Chimeric antigen receptor ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,medicine.anatomical_structure ,Cytokine ,Oncology ,Cell culture ,Immunology ,Cancer research ,Cytokine receptor ,Glioblastoma ,Signal Transduction - Abstract
Successful adoptive T-cell immunotherapy of solid tumors will require improved expansion and cytotoxicity of tumor-directed T cells within tumors. Providing recombinant or transgenic cytokines may produce the desired benefits but is associated with significant toxicities, constraining clinical use. To circumvent this limitation, we constructed a constitutively signaling cytokine receptor, C7R, which potently triggers the IL7 signaling axis but is unresponsive to extracellular cytokine. This strategy augments modified T-cell function following antigen exposure, but avoids stimulating bystander lymphocytes. Coexpressing the C7R with a tumor-directed chimeric antigen receptor (CAR) increased T-cell proliferation, survival, and antitumor activity during repeated exposure to tumor cells, without T-cell dysfunction or autonomous T-cell growth. Furthermore, C7R-coexpressing CAR T cells were active against metastatic neuroblastoma and orthotopic glioblastoma xenograft models even at cell doses that had been ineffective without C7R support. C7R may thus be able to enhance antigen-specific T-cell therapies against cancer. Significance: The constitutively signaling C7R system developed here delivers potent IL7 stimulation to CAR T cells, increasing their persistence and antitumor activity against multiple preclinical tumor models, supporting its clinical development. Cancer Discov; 7(11); 1238–47. ©2017 AACR. This article is highlighted in the In This Issue feature, p. 1201
- Published
- 2017
31. Sensing Bad: Are Co-stimulatory CAR-Expressing γδ T Cells Safer?
- Author
-
Robin Parihar
- Subjects
0301 basic medicine ,Cytotoxicity, Immunologic ,CD3 Complex ,T cell ,T-Lymphocytes ,Mutant Chimeric Proteins ,Receptors, Antigen, T-Cell ,Gene Expression ,chemical and pharmacologic phenomena ,Lymphocyte Activation ,Protein Engineering ,CD19 ,03 medical and health sciences ,Antigen ,CD28 Antigens ,Antigens, Neoplasm ,Cell Line, Tumor ,Gangliosides ,Drug Discovery ,Genetics ,medicine ,Humans ,Receptor ,Molecular Biology ,B cell ,Pharmacology ,Neurons ,biology ,Receptors, Antigen, T-Cell, gamma-delta ,Chimeric antigen receptor ,Coculture Techniques ,030104 developmental biology ,medicine.anatomical_structure ,Ectodomain ,Immunology ,Toxicity ,biology.protein ,Commentary ,Molecular Medicine ,Original Article ,Immunotherapy - Abstract
Chimeric antigen receptors (CARs) combine T cell activation with antibody-mediated tumor antigen specificity, bypassing the need for T cell receptor (TCR) ligation. A limitation of CAR technology is on-target off-tumor toxicity caused by target antigen expression on normal cells. Using GD2 as a model cancer antigen, we hypothesized that this could be minimized by using T cells expressing Vγ9Vδ2 TCR, which recognizes transformed cells in a major histocompatibility complex (MHC)-unrestricted manner, in combination with a co-stimulatory CAR that would function independently of the TCR. An anti-GD2 CAR containing a solitary endodomain derived from the NKG2D adaptor DAP10 was expressed in Vγ9Vδ2+ T cells. Differential ligation of the CAR and/or TCR using antibody-coated beads showed that pro-inflammatory cytokine response depended on activation of both receptors. Moreover, in killing assays, GD2-expressing neuroblastoma cells that engaged the Vγ9Vδ2 TCR were efficiently lysed, whereas cells that expressed GD2 equivalently but did not engage the Vγ9Vδ2 TCR were untouched. Differentiation between X-on tumor and X-off tumor offers potential for safer immunotherapy and broader target selection.
- Published
- 2017
32. 31st Annual Meeting and Associated Programs of the Society for Immunotherapy of Cancer (SITC 2016): part one
- Author
-
Andreas Lundqvist, Vincent van Hoef, Xiaonan Zhang, Erik Wennerberg, Julie Lorent, Kristina Witt, Laia Masvidal Sanz, Shuo Liang, Shannon Murray, Ola Larsson, Rolf Kiessling, Yumeng Mao, John-William Sidhom, Catherine A. Bessell, Jonathan Havel, Jonathan Schneck, Timothy A. Chan, Eliot Sachsenmeier, David Woods, Anders Berglund, Rupal Ramakrishnan, Andressa Sodre, Jeffrey Weber, Roberta Zappasodi, Yanyun Li, Jingjing Qi, Philip Wong, Cynthia Sirard, Michael Postow, Walter Newman, Henry Koon, Vamsidhar Velcheti, Margaret K. Callahan, Jedd D. Wolchok, Taha Merghoub, Lawrence G. Lum, Minsig Choi, Archana Thakur, Abhinav Deol, Gregory Dyson, Anthony Shields, Cara Haymaker, Marc Uemura, Ravi Murthy, Marihella James, Daqing Wang, Julie Brevard, Catherine Monaghan, Suzanne Swann, James Geib, Mark Cornfeld, Srinivas Chunduru, Sudhir Agrawal, Cassian Yee, Jennifer Wargo, Sapna P. Patel, Rodabe Amaria, Hussein Tawbi, Isabella Glitza, Scott Woodman, Wen-Jen Hwu, Michael A. Davies, Patrick Hwu, Willem W. Overwijk, Chantale Bernatchez, Adi Diab, Erminia Massarelli, Neil H. Segal, Vincent Ribrag, Ignacio Melero, Tara C. Gangadhar, Walter Urba, Dirk Schadendorf, Robert L. Ferris, Roch Houot, Franck Morschhauser, Theodore Logan, Jason J. Luke, William Sharfman, Fabrice Barlesi, Patrick A. Ott, Laura Mansi, Shivaani Kummar, Gilles Salles, Cecilia Carpio, Roland Meier, Suba Krishnan, Dan McDonald, Matthew Maurer, Xuemin Gu, Jaclyn Neely, Satyendra Suryawanshi, Ronald Levy, Nikhil Khushalani, Jennifer Wu, Jinyu Zhang, Fahmin Basher, Mark Rubinstein, Mark Bucsek, Guanxi Qiao, Cameron MacDonald, Bonnie Hylander, Elizabeth Repasky, Shilpak Chatterjee, Anusara Daenthanasanmak, Paramita Chakraborty, Kyle Toth, Megan Meek, Elizabeth Garrett-Mayer, Michael Nishimura, Chrystal Paulos, Craig Beeson, Xuezhong Yu, Shikhar Mehrotra, Fei Zhao, Kathy Evans, Christine Xiao, Alisha Holtzhausen, Brent A. Hanks, Nicole Scharping, Ashley V. Menk, Rebecca Moreci, Ryan Whetstone, Rebekah Dadey, Simon Watkins, Robert Ferris, Greg M. Delgoffe, Jonathan Peled, Sean Devlin, Anna Staffas, Melissa Lumish, Kori Porosnicu Rodriguez, Katya Ahr, Miguel Perales, Sergio Giralt, Ying Taur, Eric Pamer, Marcel R. M. van den Brink, Robert Jenq, Nicola Annels, Hardev Pandha, Guy Simpson, Hugh Mostafid, Kevin Harrington, Alan Melcher, Mark Grose, Bronwyn Davies, Gough Au, Roberta Karpathy, Darren Shafren, Jacob Ricca, Dmitriy Zamarin, Luciana Batista, Florence Marliot, Angela Vasaturo, Sabrina Carpentier, Cécile Poggionovo, Véronique Frayssinet, Jacques Fieschi, Marc Van den Eynde, Franck Pagès, Jérôme Galon, Fabienne Hermitte, Sean G. Smith, Khue Nguyen, Sruthi Ravindranathan, Bhanu Koppolu, David Zaharoff, Gustavo Schvartsman, Roland Bassett, Jennifer L. McQuade, Lauren E. Haydu, Douglas Kline, Xiufen Chen, Dominick Fosco, Justin Kline, Abigail Overacre, Maria Chikina, Erin Brunazzi, Gulidanna Shayan, William Horne, Jay Kolls, Tullia C. Bruno, Creg Workman, Dario Vignali, Prasad S. Adusumilli, Ephraim A Ansa-Addo, Zihai Li, Andrew Gerry, Joseph P. Sanderson, Karen Howe, Roslin Docta, Qian Gao, Eleanor A. L. Bagg, Nicholas Tribble, Miguel Maroto, Gareth Betts, Natalie Bath, Luca Melchiori, Daniel E. Lowther, Indu Ramachandran, Gabor Kari, Samik Basu, Gwendolyn Binder-Scholl, Karen Chagin, Lini Pandite, Tom Holdich, Rafael Amado, Hua Zhang, John Glod, Donna Bernstein, Bent Jakobsen, Crystal Mackall, Ryan Wong, Jonathan D. Silk, Katherine Adams, Garth Hamilton, Alan D. Bennett, Sara Brett, Junping Jing, Adriano Quattrini, Manoj Saini, Guy Wiedermann, Joanna Brewer, MyLinh Duong, An Lu, Peter Chang, Aruna Mahendravada, Nicholas Shinners, Kevin Slawin, David M. Spencer, Aaron E. Foster, J. Henri Bayle, Cristina Bergamaschi, Sinnie Sin Man Ng, Bethany Nagy, Shawn Jensen, Xintao Hu, Candido Alicea, Bernard Fox, Barbara Felber, George Pavlakis, Jessica Chacon, Tori Yamamoto, Thomas Garrabrant, Luis Cortina, Daniel J. Powell, Marco Donia, Julie Westerlin Kjeldsen, Rikke Andersen, Marie Christine Wulff Westergaard, Valentina Bianchi, Mateusz Legut, Meriem Attaf, Garry Dolton, Barbara Szomolay, Sascha Ott, Rikke Lyngaa, Sine Reker Hadrup, Andrew Kelvin Sewell, Inge Marie Svane, Aaron Fan, Takumi Kumai, Esteban Celis, Ian Frank, Amanda Stramer, Michelle A. Blaskovich, Seth Wardell, Maria Fardis, James Bender, Michael T. Lotze, Stephanie L. Goff, Nikolaos Zacharakis, Yasmine Assadipour, Todd D. Prickett, Jared J. Gartner, Robert Somerville, Mary Black, Hui Xu, Harshini Chinnasamy, Isaac Kriley, Lily Lu, John Wunderlich, Paul F. Robbins, Steven Rosenberg, Steven A. Feldman, Kasia Trebska-McGowan, Parisa Malekzadeh, Eden Payabyab, Richard Sherry, Aishwarya Gokuldass, Charlene Kopits, Brian Rabinovich, Daniel S. Green, Olena Kamenyeva, Kathryn C. Zoon, Christina M. Annunziata, Joanne Hammill, Christopher Helsen, Craig Aarts, Jonathan Bramson, Yui Harada, Yoshikazu Yonemitsu, Kenneth Mwawasi, Galina Denisova, Rajanish Giri, Benjamin Jin, Tracy Campbell, Lindsey M. Draper, Sanja Stevanovic, Zhiya Yu, Bianca Weissbrich, Nicholas P. Restifo, Cornelia L. Trimble, Christian S. Hinrichs, Kwong Tsang, Massimo Fantini, James W. Hodge, Rika Fujii, Ingrid Fernando, Caroline Jochems, Christopher Heery, James Gulley, Patrick Soon-Shiong, Jeffrey Schlom, Weiqing Jing, Jill Gershan, Grace Blitzer, James Weber, Laura McOlash, Bryon D. Johnson, Simin Kiany, Huang Gangxiong, Eugenie S. Kleinerman, Michael Klichinsky, Marco Ruella, Olga Shestova, Saad Kenderian, Miriam Kim, John Scholler, Carl H. June, Saar Gill, Duane Moogk, Shi Zhong, Ivan Liadi, William Rittase, Victoria Fang, Janna Dougherty, Arianne Perez-Garcia, Iman Osman, Cheng Zhu, Navin Varadarajan, Alan Frey, Michelle Krogsgaard, Daniel Landi, Kristen Fousek, Malini Mukherjee, Ankita Shree, Sujith Joseph, Kevin Bielamowicz, Tiara Byrd, Nabil Ahmed, Meenakshi Hegde, Sylvia Lee, David Byrd, John Thompson, Shailender Bhatia, Scott Tykodi, Judy Delismon, Liz Chu, Siddiq Abdul-Alim, Arpy Ohanian, Anna Marie DeVito, Stanley Riddell, Kim Margolin, Isabelle Magalhaes, Jonas Mattsson, Michael Uhlin, Satoshi Nemoto, Patricio Pérez Villarroel, Ryosuke Nakagawa, James J. Mule, Adam W. Mailloux, Melinda Mata, Phuong Nguyen, Claudia Gerken, Christopher DeRenzo, Stephen Gottschalk, Mélissa Mathieu, Sandy Pelletier, John Stagg, Simon Turcotte, Nicholas Minutolo, Prannda Sharma, Andrew Tsourkas, Nadine Mockel-Tenbrinck, Daniela Mauer, Katharina Drechsel, Carola Barth, Katharina Freese, Ulrike Kolrep, Silke Schult, Mario Assenmacher, Andrew Kaiser, John Mullinax, MacLean Hall, Julie Le, Krithika Kodumudi, Erica Royster, Allison Richards, Ricardo Gonzalez, Amod Sarnaik, Shari Pilon-Thomas, Morten Nielsen, Anders Krarup-Hansen, Dorrit Hovgaard, Michael Mørk Petersen, Anand Chainsukh Loya, Niels Junker, Charlotte Rivas, Robin Parihar, Cliona M. Rooney, Haiying Qin, Sang Nguyen, Paul Su, Chad Burk, Brynn Duncan, Bong-Hyun Kim, M. Eric Kohler, Terry Fry, Arjun A. Rao, Noam Teyssier, Jacob Pfeil, Nikolaos Sgourakis, Sofie Salama, David Haussler, Sarah A. Richman, Selene Nunez-Cruz, Zack Gershenson, Zissimos Mourelatos, David Barrett, Stephan Grupp, Michael Milone, Alba Rodriguez-Garcia, Matthew K. Robinson, Gregory P. Adams, João Santos, Riikka Havunen, Mikko Siurala, Víctor Cervera-Carrascón, Suvi Parviainen, Marjukka Antilla, Akseli Hemminki, Jyothi Sethuraman, Laurelis Santiago, Jie Qing Chen, Zhimin Dai, Huizi Sha, Shu Su, Naiqing Ding, Baorui Liu, Anna Pasetto, Sarah R. Helman, Steven A. Rosenberg, Melissa Burgess, Hui Zhang, Tien Lee, Hans Klingemann, Paul Nghiem, John M. Kirkwood, John M. Rossi, Marika Sherman, Allen Xue, Yueh-wei Shen, Lynn Navale, James N. Kochenderfer, Adrian Bot, Anandaraman Veerapathran, Doris Wiener, Edmund K. Waller, Jian-Ming Li, Christopher Petersen, Bruce R. Blazar, Jingxia Li, Cynthia R. Giver, Ziming Wang, Steven K. Grossenbacher, Ian Sturgill, Robert J. Canter, William J. Murphy, Congcong Zhang, Michael C. Burger, Lukas Jennewein, Anja Waldmann, Michel Mittelbronn, Torsten Tonn, Joachim P. Steinbach, Winfried S. Wels, Jason B. Williams, Yuanyuan Zha, Thomas F. Gajewski, LaTerrica C. Williams, Giedre Krenciute, Mamta Kalra, Chrystal Louis, Gang Xin, David Schauder, Aimin Jiang, Nikhil Joshi, Weiguo Cui, Xue Zeng, Zeguo Zhao, Mohamad Hamieh, Justin Eyquem, Gertrude Gunset, Neil Bander, Michel Sadelain, David Askmyr, Milad Abolhalaj, Kristina Lundberg, Lennart Greiff, Malin Lindstedt, Helen K. Angell, Kyoung-Mee Kim, Seung-Tae Kim, Sung Kim, Alan D. Sharpe, Julia Ogden, Anna Davenport, Darren R. Hodgson, Carl Barrett, Jeeyun Lee, Elaine Kilgour, Jodi Hanson, Richard Caspell, Alexey Karulin, Paul Lehmann, Tameem Ansari, Annemarie Schiller, Srividya Sundararaman, Diana Roen, Mark Ayers, Diane Levitan, Gladys Arreaza, Fang Liu, Robin Mogg, Yung-Jue Bang, Bert O’Neil, Razvan Cristescu, Philip Friedlander, Karl Wassman, Chrisann Kyi, William Oh, Nina Bhardwaj, Svetlana Bornschlegl, Michael P. Gustafson, Dennis A. Gastineau, Ian F. Parney, Allan B. Dietz, Daniel Carvajal-Hausdorf, Nikita Mani, Kurt Schalper, David Rimm, Serena Chang, John Kurland, Christoph Matthias Ahlers, Maria Jure-Kunkel, Lewis Cohen, Holden Maecker, Holbrook Kohrt, Shuming Chen, George Crabill, Theresa Pritchard, Tracee McMiller, Drew Pardoll, Fan Pan, Suzanne Topalian, Patrick Danaher, Sarah Warren, Lucas Dennis, Andrew M. White, Leonard D’Amico, Melissa Geller, Mary L. Disis, Joseph Beechem, Kunle Odunsi, Steven Fling, Roshanak Derakhshandeh, Tonya J. Webb, Sigrid Dubois, Kevin Conlon, Bonita Bryant, Jennifer Hsu, Nancy Beltran, Jürgen Müller, Thomas Waldmann, Rebekka Duhen, Thomas Duhen, Lucas Thompson, Ryan Montler, Andrew Weinberg, Max Kates, Brandon Early, Erik Yusko, Taylor H. Schreiber, Trinity J. Bivalacqua, Jared Lunceford, Michael Nebozhyn, Erin Murphy, Andrey Loboda, David R. Kaufman, Andrew Albright, Jonathan Cheng, S. Peter Kang, Veena Shankaran, Sarina A. Piha-Paul, Jennifer Yearley, Tanguy Seiwert, Antoni Ribas, Terrill K. McClanahan, Xinwei Sher, Xiao Qiao Liu, Andrew Joe, Elizabeth Plimack, Alex Forrest-Hay, Cheryl A. Guyre, Kohei Narumiya, Marc Delcommenne, Heather A. Hirsch, Amit Deshpande, Jason Reeves, Jenny Shu, Tong Zi, Jennifer Michaelson, Debbie Law, Elizabeth Trehu, Sriram Sathyanaryanan, Brendan P. Hodkinson, Natalie A. Hutnick, Michael E. Schaffer, Michael Gormley, Tyler Hulett, Carmen Ballesteros-Merino, Christopher Dubay, Michael Afentoulis, Ashok Reddy, Larry David, Kumar Jayant, Swati Agrawal, Rajendra Agrawal, Ghayathri Jeyakumar, Seongho Kim, Heejin Kim, Cynthia Silski, Stacey Suisham, Elisabeth Heath, Ulka Vaishampayan, Natalie Vandeven, Natasja Nielsen Viller, Alison O’Connor, Hui Chen, Bolette Bossen, Eric Sievers, Robert Uger, Lisa Johnson, Hsiang-Fong Kao, Chin-Fu Hsiao, Shu-Chuan Lai, Chun-Wei Wang, Jenq-Yuh Ko, Pei-Jen Lou, Tsai-Jan Lee, Tsang-Wu Liu, Ruey-Long Hong, Staci J. Kearney, Joshua C. Black, Benjamin J. Landis, Sally Koegler, Brooke Hirsch, Roberto Gianani, Jeffrey Kim, Ming-Xiao He, Bingqing Zhang, Nan Su, Yuling Luo, Xiao-Jun Ma, Emily Park, Dae Won Kim, Domenico Copploa, Nishi Kothari, Young doo Chang, Richard Kim, Namyong Kim, Melvin Lye, Ee Wan, Hanna A. Knaus, Sofia Berglund, Hubert Hackl, Judith E. Karp, Ivana Gojo, Leo Luznik, Henoch S. Hong, Sven D. Koch, Birgit Scheel, Ulrike Gnad-Vogt, Karl-Josef Kallen, Volker Wiegand, Linus Backert, Oliver Kohlbacher, Ingmar Hoerr, Mariola Fotin-Mleczek, James M. Billingsley, Yoshinobu Koguchi, Valerie Conrad, William Miller, Iliana Gonzalez, Tomasz Poplonski, Tanisha Meeuwsen, Ana Howells-Ferreira, Rogan Rattray, Mary Campbell, Carlo Bifulco, Keith Bahjat, Brendan Curti, E-K Vetsika, G. Kallergi, Despoina Aggouraki, Z. Lyristi, P. Katsarlinos, Filippos Koinis, V. Georgoulias, Athanasios Kotsakis, Nathan T. Martin, Famke Aeffner, Logan Cerkovnik, Luke Pratte, Rebecca Kim, Joseph Krueger, Amaia Martínez-Usatorre, Camilla Jandus, Alena Donda, Laura Carretero-Iglesia, Daniel E. Speiser, Dietmar Zehn, Nathalie Rufer, Pedro Romero, Anshuman Panda, Janice Mehnert, Kim M. Hirshfield, Greg Riedlinger, Sherri Damare, Tracie Saunders, Levi Sokol, Mark Stein, Elizabeth Poplin, Lorna Rodriguez-Rodriguez, Ann Silk, Nancy Chan, Melissa Frankel, Michael Kane, Jyoti Malhotra, Joseph Aisner, Howard L. Kaufman, Siraj Ali, Jeffrey Ross, Eileen White, Gyan Bhanot, Shridar Ganesan, Anne Monette, Derek Bergeron, Amira Ben Amor, Liliane Meunier, Christine Caron, Antigoni Morou, Daniel Kaufmann, Moishe Liberman, Igor Jurisica, Anne-Marie Mes-Masson, Kamel Hamzaoui, Rejean Lapointe, Ann Mongan, Yuan-Chieh Ku, Warren Tom, Yongming Sun, Alex Pankov, Tim Looney, Janice Au-Young, Fiona Hyland, Jeff Conroy, Carl Morrison, Sean Glenn, Blake Burgher, He Ji, Mark Gardner, Angela R. Omilian, Wiam Bshara, Omilian Angela, Joseph M. Obeid, Gulsun Erdag, Mark E. Smolkin, Donna H. Deacon, James W. Patterson, Lieping Chen, Timothy N. Bullock, Craig L. Slingluff, John T. Loffredo, Raja Vuyyuru, Sophie Beyer, Vanessa M. Spires, Maxine Fox, Jon M. Ehrmann, Katrina A. Taylor, Alan J. Korman, Robert F. Graziano, David Page, Katherine Sanchez, Maritza Martel, Mariana Petaccia De Macedo, Yong Qin, Alex Reuben, Christine Spencer, Michele Guindani, Adriana Racolta, Brian Kelly, Tobin Jones, Nathan Polaske, Noah Theiss, Mark Robida, Jeffrey Meridew, Iva Habensus, Liping Zhang, Lidija Pestic-Dragovich, Lei Tang, Ryan J. Sullivan, Thomas Olencki, Thomas Hutson, Joanna Roder, Shauna Blackmon, Heinrich Roder, John Stewart, Asim Amin, Marc S. Ernstoff, Joseph I. Clark, Michael B. Atkins, Jeffrey Sosman, David F. McDermott, Harriet Kluger, Ruth Halaban, Mario Snzol, Senait Asmellash, Arni Steingrimsson, Chichung Wang, Kristin Roman, Amanda Clement, Sean Downing, Clifford Hoyt, Nathalie Harder, Guenter Schmidt, Ralf Schoenmeyer, Nicolas Brieu, Mehmet Yigitsoy, Gabriele Madonna, Gerardo Botti, Antonio Grimaldi, Paolo A. Ascierto, Ralf Huss, Maria Athelogou, Harald Hessel, Alexander Buchner, Christian Stief, Gerd Binnig, Thomas Kirchner, Shankar Sellappan, Sheeno Thyparambil, Sarit Schwartz, Fabiola Cecchi, Andrew Nguyen, Charles Vaske, Todd Hembrough, Jan Spacek, Michal Vocka, Eva Zavadova, Helena Skalova, Pavel Dundr, Lubos Petruzelka, Nicole Francis, Rau T. Tilman, Arndt Hartmann, Irena Netikova, Julia Stump, Amanda Tufman, Frank Berger, Michael Neuberger, Rudolf Hatz, Michael Lindner, Rachel E. Sanborn, John Handy, Rudolf M. Huber, Hauke Winter, Simone Reu, Cheng Sun, Weihua Xiao, Zhigang Tian, Kshitij Arora, Niyati Desai, Anupriya Kulkarni, Mihir Rajurkar, Miguel Rivera, Vikram Deshpande, David Ting, Katy Tsai, Adi Nosrati, Simone Goldinger, Omid Hamid, Alain Algazi, Paul Tumeh, Jimmy Hwang, Jacqueline Liu, Lawrence Chen, Reinhard Dummer, Michael Rosenblum, Adil Daud, Tsu-Shuen Tsao, Julia Ashworth-Sharpe, Donald Johnson, Srabani Bhaumik, Christopher Bieniarz, Joseph Couto, Michael Farrell, Mahsa Ghaffari, Antony Hubbard, Jerome Kosmeder, Cleo Lee, Erin Marner, Diana Uribe, Hongjun Zhang, Jian Zhang, Wenjun Zhang, Yifei Zhu, Larry Morrison, Takahiro Tsujikawa, Rohan N. Borkar, Vahid Azimi, Sushil Kumar, Guillaume Thibault, Motomi Mori, Edward El Rassi, Daniel R. Clayburgh, Molly F. Kulesz-Martin, Paul W. Flint, Lisa M. Coussens, Lisa Villabona, Giuseppe V. Masucci, Gary Geiss, Brian Birditt, Qian Mei, Alan Huang, Maribeth A. Eagan, Eduardo Ignacio, Nathan Elliott, Dwayne Dunaway, Jaemyeong Jung, Chris Merritt, Isaac Sprague, Philippa Webster, Yan Liang, Jessica Wenthe, Gunilla Enblad, Hannah Karlsson, Magnus Essand, Barbara Savoldo, Gianpietro Dotti, Martin Höglund, Malcolm K. Brenner, Hans Hagberg, Angelica Loskog, Matthew J. Bernett, Gregory L. Moore, Michael Hedvat, Christine Bonzon, Seung Chu, Rumana Rashid, Kendra N. Avery, Umesh Muchhal, John Desjarlais, Matthew Kraman, Katarzyna Kmiecik, Natalie Allen, Mustapha Faroudi, Carlo Zimarino, Mateusz Wydro, Jacqueline Doody, Sreesha P. Srinivasa, Nagaraja Govindappa, Praveen Reddy, Aparajita Dubey, Sankar Periyasamy, Madhukara Adekandi, Chaitali Dey, Mary Joy, Pieter Fokko van Loo, Henrike Veninga, Setareh Shamsili, Mark Throsby, Harry Dolstra, Lex Bakker, Ajjai Alva, Juergen Gschwendt, Yohann Loriot, Joaquim Bellmunt, Dai Feng, Christian Poehlein, Thomas Powles, Emmanuel S. Antonarakis, Charles G. Drake, Haiyan Wu, Johann De Bono, Rajat Bannerji, John Byrd, Gareth Gregory, Stephen Opat, Jake Shortt, Andrew J. Yee, Noopur Raje, Seth Thompson, Arun Balakumaran, Shaji Kumar, Brian I. Rini, Toni K. Choueiri, Mariangela Mariani, Laurence Albiges, John B. Haanen, James Larkin, Manuela Schmidinger, Domenico Magazzù, Alessandra di Pietro, Robert J. Motzer, Troels Holz Borch, Per Kongsted, Magnus Pedersen, Özcan Met, Karim Boudadi, Hao Wang, James Vasselli, Jan E. Baughman, Jon Wigginton, Rehab Abdallah, Ashley Ross, Jiwon Park, Steven Grossenbacher, Jesus I. Luna, Sita Withers, William Culp, Mingyi Chen, Arta Monjazeb, Michael S. Kent, Smita Chandran, David Danforth, James Yang, Christopher Klebanoff, Stephanie Goff, Biman Paria, Arvind Sabesan, Abhishek Srivastava, Udai Kammula, Jon Richards, Mark Faries, Robert H. I. Andtbacka, Luis A. Diaz, Dung T. Le, Takayuki Yoshino, Thierry André, Johanna Bendell, Minori Koshiji, Yayan Zhang, S Peter Kang, Bao Lam, Dirk Jäger, Todd M. Bauer, Judy S. Wang, Jean K. Lee, Gulam A. Manji, Ragini Kudchadkar, John S. Kauh, Shande Tang, Naomi Laing, Gerald Falchook, Edward B. Garon, Balazs Halmos, Hui Rina, Natasha Leighl, Sung Sook Lee, William Walsh, Konstanin Dragnev, Bilal Piperdi, Luis Paz-Ares Rodriguez, Nabeegha Shinwari, Ziewn Wei, Mary L Maas, Michael Deeds, Adam Armstrong, Tim Peterson, Sue Steinmetz, Thomas Herzog, Floor J. Backes, Larry Copeland, Maria Del Pilar Estevez Diz, Thomas W. Hare, Warner Huh, Byoung-Gie Kim, Kathleen M. Moore, Ana Oaknin, William Small, Krishnansu S. Tewari, Bradley J. Monk, Ashish M. Kamat, Kijoeng Nam, Maria De Santis, Robert Dreicer, Noah M. Hahn, Rodolfo Perini, Arlene Siefker-Radtke, Guru Sonpavde, Ronald de Wit, J. Alfred Witjes, Stephen Keefe, Dean Bajorin, Philippe Armand, John Kuruvilla, Craig Moskowitz, Mehdi Hamadani, Pier Luigi Zinzani, Sabine Chlosta, Nancy Bartlett, Rachel Sabado, Yvonne Saenger, Loging William, Michael Joseph Donovan, Erlinda Sacris, John Mandeli, Andres M. Salazar, John Powderly, Joshua Brody, John Nemunaitis, Leisha Emens, Amita Patnaik, Ian McCaffery, Richard Miller, Ginna Laport, Andrew L. Coveler, David C. Smith, Juneko E. Grilley-Olson, Sanjay Goel, Shyra J. Gardai, Che-Leung Law, Gary Means, Thomas Manley, Kristen A. Marrone, Gary Rosner, Valsamo Anagnostou, Joanne Riemer, Jessica Wakefield, Cynthia Zanhow, Stephen Baylin, Barbara Gitlitz, Julie Brahmer, Sabina Signoretti, Wenting Li, Charles Schloss, Jean-Marie Michot, Wei Ding, Beth Christian, Patricia Marinello, Margaret Shipp, Yana G. Najjar, null Lin, Lisa H. Butterfield, Ahmad A. Tarhini, Diwakar Davar, Hassane Zarour, Elizabeth Rush, Cindy Sander, Siqing Fu, Todd Bauer, Chris Molineaux, Mark K. Bennett, Keith W. Orford, Kyriakos P. Papadopoulos, Sukhmani K. Padda, Sumit A. Shah, A Dimitrios Colevas, Sujata Narayanan, George A. Fisher, Dana Supan, Heather A. Wakelee, Rhonda Aoki, Mark D. Pegram, Victor M. Villalobos, Jie Liu, Chris H. Takimoto, Mark Chao, Jens-Peter Volkmer, Ravindra Majeti, Irving L. Weissman, Branimir I. Sikic, Wendy Yu, Alison Conlin, Janet Ruzich, Stacy Lewis, Anupama Acheson, Kathleen Kemmer, Kelly Perlewitz, Nicole M. Moxon, Staci Mellinger, Heather McArthur, Trine Juhler-Nøttrup, Jayesh Desai, Ben Markman, Shahneen Sandhu, Hui Gan, Michael L. Friedlander, Ben Tran, Tarek Meniawy, Joanne Lundy, Duncan Colyer, Malaka Ameratunga, Christie Norris, Jason Yang, Kang Li, Lai Wang, Lusong Luo, Zhen Qin, Song Mu, Xuemei Tan, James Song, Michael Millward, Matthew H. G. Katz, Todd W. Bauer, Gauri R. Varadhachary, Nicolas Acquavella, Nipun Merchant, Gina Petroni, Osama E. Rahma, Mei Chen, Yang Song, Markus Puhlmann, Arun Khattri, Ryan Brisson, Christopher Harvey, Jatin Shah, Maria Victoria Mateos, Morio Matsumoto, Hilary Blacklock, Albert Oriol Rocafiguera, Hartmut Goldschmidt, Shinsuke Iida, Dina Ben Yehuda, Enrique Ocio, Paula Rodríguez-Otero, Sundar Jagannath, Sagar Lonial, Uma Kher, Jesus San-Miguel, Moacyr Ribeiro de Oliveira, Habte Yimer, Robert Rifkin, Fredrik Schjesvold, Razi Ghori, Anna Spreafico, Victor Lee, Roger K. C. Ngan, Ka Fai To, Myung Ju Ahn, Quan Sing Ng, Jin-Ching Lin, Ramona F. Swaby, Christine Gause, Sanatan Saraf, Anthony T. C. Chan, Elaine Lam, Nizar M. Tannir, Funda Meric-Bernstam, Matt Gross, Andy MacKinnon, Sam Whiting, Martin Voss, Evan Y. Yu, Mark R. Albertini, Erik A. Ranheim, Jacquelyn A. Hank, Cindy Zuleger, Thomas McFarland, Jennifer Collins, Erin Clements, Sharon Weber, Tracey Weigel, Heather Neuman, Greg Hartig, David Mahvi, MaryBeth Henry, Jacek Gan, Richard Yang, Lakeesha Carmichael, KyungMann Kim, Stephen D. Gillies, Paul M. Sondel, Vivek Subbiah, Lori Noffsinger, Kyle Hendricks, Marnix Bosch, Jay M. Lee, Mi-Heon Lee, Jonathan W. Goldman, Felicita E. Baratelli, Dorthe Schaue, Gerald Wang, Frances Rosen, Jane Yanagawa, Tonya C. Walser, Ying Q. Lin, Sharon Adams, Franco M. Marincola, Paul C. Tumeh, Fereidoun Abtin, Robert Suh, Karen Reckamp, William D. Wallace, Gang Zeng, David A. Elashoff, Sherven Sharma, Steven M. Dubinett, Anna C. Pavlick, Brian Gastman, Brent Hanks, Tibor Keler, Tom Davis, Laura A. Vitale, Elad Sharon, Chihiro Morishima, Martin Cheever, Christopher R. Heery, Joseph W. Kim, Elizabeth Lamping, Jennifer Marte, Sheri McMahon, Lisa Cordes, Farhad Fakhrejahani, Ravi Madan, Rachel Salazar, Maggie Zhang, Christoph Helwig, James L Gulley, Roger Li, John Amrhein, Zvi Cohen, Monique Champagne, Ashish Kamat, M. Angela Aznar, Sara Labiano, Angel Diaz-Lagares, Manel Esteller, Juan Sandoval, Susannah D. Barbee, David I. Bellovin, John C. Timmer, Nebiyu Wondyfraw, Susan Johnson, Johanna Park, Amanda Chen, Mikayel Mkrtichyan, Amir S. Razai, Kyle S. Jones, Chelsie Y. Hata, Denise Gonzalez, Quinn Deveraux, Brendan P. Eckelman, Luis Borges, Rukmini Bhardwaj, Raj K. Puri, Akiko Suzuki, Pamela Leland, Bharat H. Joshi, Todd Bartkowiak, Ashvin Jaiswal, Casey Ager, Midan Ai, Pratha Budhani, Renee Chin, David Hong, Michael Curran, William D. Hastings, Maria Pinzon-Ortiz, Masato Murakami, Jason R. Dobson, David Quinn, Joel P. Wagner, Xianhui Rong, Pamela Shaw, Ernesta Dammassa, Wei Guan, Glenn Dranoff, Alexander Cao, Ross B. Fulton, Steven Leonardo, Kathryn Fraser, Takashi O. Kangas, Nadine Ottoson, Nandita Bose, Richard D. Huhn, Jeremy Graff, Jamie Lowe, Keith Gorden, Mark Uhlik, Thomas O’Neill, Jenifer Widger, Andrea Crocker, Li-Zhen He, Jeffrey Weidlick, Karuna Sundarapandiyan, Venky Ramakrishna, James Storey, Lawrence J. Thomas, Joel Goldstein, Henry C. Marsh, Jamison Grailer, Julia Gilden, Pete Stecha, Denise Garvin, Jim Hartnett, Frank Fan, Mei Cong, Zhi-jie Jey Cheng, Marlon J. Hinner, Rachida-Siham Bel Aiba, Corinna Schlosser, Thomas Jaquin, Andrea Allersdorfer, Sven Berger, Alexander Wiedenmann, Gabriele Matschiner, Julia Schüler, Ulrich Moebius, Christine Rothe, Olwill A. Shane, Brendan Horton, Stefani Spranger, Dayson Moreira, Tomasz Adamus, Xingli Zhao, Piotr Swiderski, Sumanta Pal, Marcin Kortylewski, Alyssa Kosmides, Kevin Necochea, Kathleen M. Mahoney, Sachet A. Shukla, Nikolaos Patsoukis, Apoorvi Chaudhri, Hung Pham, Ping Hua, Xia Bu, Baogong Zhu, Nir Hacohen, Catherine J. Wu, Edward Fritsch, Vassiliki A. Boussiotis, Gordon J. Freeman, Amy E. Moran, Fanny Polesso, Lisa Lukaesko, Emelie Rådestad, Lars Egevad, Berit Sundberg, Lars Henningsohn, Victor Levitsky, William Rafelson, John L. Reagan, Loren Fast, Pottayil Sasikumar, Naremaddepalli Sudarshan, Raghuveer Ramachandra, Nagesh Gowda, Dodheri Samiulla, Talapaneni Chandrasekhar, Sreenivas Adurthi, Jiju Mani, Rashmi Nair, Amit Dhudashia, Nagaraj Gowda, Murali Ramachandra, Alexander Sankin, Benjamin Gartrell, Kerwin Cumberbatch, Hongying Huang, Joshua Stern, Mark Schoenberg, Xingxing Zang, Ryan Swanson, Michael Kornacker, Lawrence Evans, Erika Rickel, Martin Wolfson, Sandrine Valsesia-Wittmann, Tala Shekarian, François Simard, Rodrigo Nailo, Aurélie Dutour, Anne-Catherine Jallas, Christophe Caux, and Aurélien Marabelle
- Subjects
Pharmacology ,0303 health sciences ,Cancer Research ,Side effect ,business.industry ,medicine.drug_class ,Immunology ,Phases of clinical research ,Monoclonal antibody ,Phase i study ,Clinical trial ,03 medical and health sciences ,0302 clinical medicine ,Oncology ,Pharmacokinetics ,030220 oncology & carcinogenesis ,Molecular Medicine ,Immunology and Allergy ,Medicine ,In patient ,Programmed death 1 ,business ,030304 developmental biology - Published
- 2016
- Full Text
- View/download PDF
33. Large-Scale Culture and Genetic Modification of Human Natural Killer Cells for Cellular Therapy
- Author
-
Natalia, Lapteva, Robin, Parihar, Lisa A, Rollins, Adrian P, Gee, and Cliona M, Rooney
- Subjects
Killer Cells, Natural ,Cell Transplantation ,Transduction, Genetic ,Blood Component Removal ,Cell Culture Techniques ,Feeder Cells ,Humans ,K562 Cells ,Lymphocyte Activation ,Cell Proliferation - Abstract
Recent advances in methods for the ex vivo expansion of human natural killer (NK) cells have facilitated the use of these powerful immune cells in clinical protocols. Further, the ability to genetically modify primary human NK cells following rapid expansion allows targeting and enhancement of their immune function. We have successfully adapted an expansion method for primary NK cells from peripheral blood mononuclear cells or from apheresis products in gas permeable rapid expansion devices (G-Rexes). Here, we describe an optimized protocol for rapid and robust NK cell expansion as well as a method for highly efficient retroviral transduction of these ex vivo expanded cells. These methodologies are good manufacturing practice (GMP) compliant and could be used for clinical-grade product manufacturing.
- Published
- 2016
34. Abstract 4734: Suppressive myeloid cells of the solid tumor microenvironment enhance regulatory T cell function and differentially affect CAR-T cell function
- Author
-
Anna Cole, Robin Parihar, Cliona M. Rooney, and Charlotte H. Rivas
- Subjects
Cancer Research ,Tumor microenvironment ,biology ,Regulatory T cell ,medicine.medical_treatment ,Immunotherapy ,CD19 ,medicine.anatomical_structure ,Immune system ,Oncology ,Antigen ,Tumor progression ,biology.protein ,Cancer research ,medicine ,Homing (hematopoietic) - Abstract
Immunotherapy with tumor-directed immune cells, such as chimeric antigen receptor-bearing T (CAR-T) cells, have shown great promise. Indeed, CAR-T cells directed against leukemia antigens such as CD19 have produced impressive complete responses even in relapsed disease patients. In contrast, CAR-T cells directed against solid tumor have produced very few durable clinical responses. This may be, in part, due to the suppressive immune microenvironment of solid tumors that inhibits the anti-tumor activity of endogenous and CAR-T cells. Myeloid-derived suppressor cells (MDSCs), regulatory T cells (Tregs) and inhibitory macrophages (M2s) are recruited by tumor-derived factors, and contribute to the immunosuppressive tumor microenvironment (TME) by secreting suppressive cytokines, expressing inhibitory ligands, and promoting tumor neovascularization. However, the contribution of each of these components to suppressing engineered cellular therapies has not been well defined. In order to design more effective cellular therapies for solid tumors, it is imperative that we understand TME-mediated immune suppression of human CAR-T cells. Therefore, the objective of this study was to evaluate the individual contribution of MDSCs, Tregs, and M2s to the suppressive TME and their ability to modulate CAR-T function. We hypothesized that MDSCs, Tregs, and M2s would inhibit GD2.CAR-T homing and proliferation at tumor sites, leading to decreased activity and tumor progression. To understand the individual contributions of these suppressive cells to inhibition of CAR-T cells, we developed a unique TME model in which human neuroblastoma tumor cells were co-inoculated subcutaneously with MDSCs, Tregs, or M2s in immunocompromised mice and then treated with GD2 neuroblastoma antigen-specific CAR-T cells. We found that while all three cell types decreased GD2.CAR-T cell-mediated tumor control in vivo, only MDSCs and M2s inhibited CAR-T homing and expansion at tumor sites. To investigate cross-talk between these suppressive cells, we assessed the phenotype and suppressive activity of MDSCs, Tregs, and M2 on autologous CAR-T cells after exposure to the other suppressive cell types. We found that the suppressive function of M2s increased by 30% after exposure to Tregs. Similarly, Treg suppressive function increased by 26% after exposure to M2s. Both M2 and Treg suppressive functions were enhanced in the presence of MDSCs. Our results suggest a potential cross-talk between suppressive cells of the TME and distinct roles in important aspects of CAR-T cell function, including homing and killing. Future studies in our lab aim to further understand the differential roles of these cells in regulating CAR-T cell function within the TME that will inform new approaches to engineer effective adoptive T-cell therapies. Citation Format: Charlotte H. Rivas, Anna Cole, Cliona M. Rooney, Robin Parihar. Suppressive myeloid cells of the solid tumor microenvironment enhance regulatory T cell function and differentially affect CAR-T cell function [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 4734.
- Published
- 2018
- Full Text
- View/download PDF
35. Colocalization of the IL-12 receptor and FcγRIIIa to natural killer cell lipid rafts leads to activation of ERK and enhanced production of interferon-γ
- Author
-
Sri Vidya Kondadasula, William E. Carson, Michael A. Caligiuri, Jianhua Yu, Susheela Tridandapani, Robin Parihar, Julie M. Roda, Amy Lehman, and Richard W. Burry
- Subjects
MAPK/ERK pathway ,Immunology ,Syk ,Cell Biology ,Hematology ,Transfection ,Biology ,Biochemistry ,Natural killer cell ,Cell biology ,medicine.anatomical_structure ,medicine ,Interleukin 12 ,Interferon gamma ,Receptor ,Lipid raft ,medicine.drug - Abstract
Natural killer (NK) cells express an activating receptor for the Fc portion of IgG (FcγRIIIa) that mediates interferon (IFN)–γ production in response to antibody (Ab)–coated targets. We have previously demonstrated that NK cells activated with interleukin-12 (IL-12) in the presence of immobilized IgG secrete 10-fold or more higher levels of IFN-γ as compared with stimulation with either agent alone. We examined the intracellular signaling pathways responsible for this synergistic IFN-γ production. NK cells costimulated via the FcR and the IL-12 receptor (IL-12R) exhibited enhanced levels of activated STAT4 and Syk as compared with NK cells stimulated through either receptor alone. Extracellular signal–regulated kinase (ERK) was also synergistically activated under these conditions. Studies with specific chemical inhibitors revealed that the activation of ERK was dependent on the activation of PI3-K, whose activation was dependent on Syk, and that sequential activation of these molecules was required for NK cell IFN-γ production in response to FcR and IL-12 stimulation. Retroviral transfection of ERK1 into primary human NK cells substantially increased IFN-γ production in response to immobilized IgG and IL-12, while transfection of human NK cells with a dominant-negative ERK1 abrogated IFN-γ production. Confocal microscopy and cellular fractionation experiments revealed that FcγRIIIa and the IL-12R colocalized to areas of lipid raft microdomains in response to costimulation with IgG and IL-12. Chemical disruption of lipid rafts inhibited ERK signaling in response to costimulation and significantly inhibited IFN-γ production. These data suggest that dual recruitment of FcγRIIIa and the IL-12R to lipid raft microdomains allows for enhanced activation of downstream signaling events that lead to IFN-γ production.
- Published
- 2008
- Full Text
- View/download PDF
36. Large-Scale Culture and Genetic Modification of Human Natural Killer Cells for Cellular Therapy
- Author
-
Cliona M. Rooney, Natalia Lapteva, Adrian P. Gee, Robin Parihar, and Lisa Rollins
- Subjects
0301 basic medicine ,Cell therapy ,03 medical and health sciences ,030104 developmental biology ,Immune system ,Rapid expansion ,Good manufacturing practice ,Ex vivo expansion ,Biology ,Retroviral transduction ,Peripheral blood mononuclear cell ,Ex vivo ,Cell biology - Abstract
Recent advances in methods for the ex vivo expansion of human natural killer (NK) cells have facilitated the use of these powerful immune cells in clinical protocols. Further, the ability to genetically modify primary human NK cells following rapid expansion allows targeting and enhancement of their immune function. We have successfully adapted an expansion method for primary NK cells from peripheral blood mononuclear cells or from apheresis products in gas permeable rapid expansion devices (G-Rexes). Here, we describe an optimized protocol for rapid and robust NK cell expansion as well as a method for highly efficient retroviral transduction of these ex vivo expanded cells. These methodologies are good manufacturing practice (GMP) compliant and could be used for clinical-grade product manufacturing.
- Published
- 2016
- Full Text
- View/download PDF
37. Gene expression profiling of the human natural killer cell response to Fc receptor activation: unique enhancement in the presence of interleukin-12
- Author
-
Andrew Stiff, William E. Carson, Omkar Lele, Steven D. Scoville, Neela Bhave, Ramana V. Davuluri, Sri Vidya Kondadasula, Amanda Campbell, Kelly Regan, Sandya Liyanarachchi, Philip R. O. Payne, Arka Pattanayak, Prashant Trikha, and Robin Parihar
- Subjects
Cell signaling ,Fc receptor ,Receptors, Fc ,NK cells ,CD16 ,Biology ,Natural killer cell ,Interferon-gamma ,Genetics ,medicine ,Humans ,Genetics(clinical) ,Gene Regulatory Networks ,Gene microarray ,Genetics (clinical) ,Oligonucleotide Array Sequence Analysis ,Genomics ,Interleukin-12 ,3. Good health ,Cell biology ,Killer Cells, Natural ,Gene expression profiling ,medicine.anatomical_structure ,Immunoglobulin G ,biology.protein ,Interleukin 12 ,Cytokine secretion ,Signal transduction ,Transcriptome ,Research Article - Abstract
Background Traditionally, the CD56dimCD16+ subset of Natural Killer (NK) cells has been thought to mediate cellular cytotoxicity with modest cytokine secretion capacity. However, studies have suggested that this subset may exert a more diverse array of immunological functions. There exists a lack of well-developed functional models to describe the behavior of activated NK cells, and the interactions between signaling pathways that facilitate effector functions are not well understood. In the present study, a combination of genome-wide microarray analyses and systems-level bioinformatics approaches were utilized to elucidate the transcriptional landscape of NK cells activated via interactions with antibody-coated targets in the presence of interleukin-12 (IL-12). Methods We conducted differential gene expression analysis of CD56dimCD16+ NK cells following FcR stimulation in the presence or absence of IL-12. Next, we functionally characterized gene sets according to patterns of gene expression and validated representative genes using RT-PCR. IPA was utilized for biological pathway analysis, and an enriched network of interacting genes was generated using GeneMANIA. Furthermore, PAJEK and the HITS algorithm were employed to identify important genes in the network according to betweeness centrality, hub, and authority node metrics. Results Analyses revealed that CD56dimCD16+ NK cells co-stimulated via the Fc receptor (FcR) and IL-12R led to the expression of a unique set of genes, including genes encoding cytotoxicity receptors, apoptotic proteins, intracellular signaling molecules, and cytokines that may mediate enhanced cytotoxicity and interactions with other immune cells within inflammatory tissues. Network analyses identified a novel set of connected key players, BATF, IRF4, TBX21, and IFNG, within an integrated network composed of differentially expressed genes in NK cells stimulated by various conditions (immobilized IgG, IL-12, or the combination of IgG and IL-12). Conclusions These results are the first to address the global mechanisms by which NK cells mediate their biological functions when encountering antibody-coated targets within inflammatory sites. Moreover, this study has identified a set of high-priority targets for subsequent investigation into strategies to combat cancer by enhancing the anti-tumor activity of CD56dimCD16+ NK cells. Electronic supplementary material The online version of this article (doi:10.1186/s12920-015-0142-9) contains supplementary material, which is available to authorized users.
- Published
- 2015
- Full Text
- View/download PDF
38. Interleukin-21 Enhances NK Cell Activation in Response to Antibody-Coated Targets
- Author
-
William E. Carson, Amy Lehman, Susheela Tridandapani, Aruna Mani, Robin Parihar, and Julie M. Roda
- Subjects
Chemokine ,Receptor, ErbB-2 ,Immunology ,Antineoplastic Agents ,Breast Neoplasms ,Biology ,Antibodies, Monoclonal, Humanized ,Lymphocyte Activation ,CD49b ,Interferon-gamma ,Mice ,Interleukin 21 ,Immune system ,Adjuvants, Immunologic ,T-Lymphocyte Subsets ,Cell Line, Tumor ,Animals ,Humans ,Immunology and Allergy ,Mice, Inbred BALB C ,Lymphokine-activated killer cell ,Tumor Necrosis Factor-alpha ,Interleukins ,Janus kinase 3 ,Antibodies, Monoclonal ,Trastuzumab ,Cytotoxicity Tests, Immunologic ,Killer Cells, Natural ,Chemotaxis, Leukocyte ,Disease Models, Animal ,Immunoglobulin G ,Cancer cell ,biology.protein ,Cancer research ,Interleukin 12 ,Female ,Chemokines ,Neoplasm Transplantation - Abstract
NK cells express an activating FcR (FcγRIIIa) that mediates Ab-dependent cellular cytotoxicity and the production of immune modulatory cytokines in response to Ab-coated targets. IL-21 has antitumor activity in murine models that depends in part on its ability to promote NK cell cytotoxicity and IFN-γ secretion. We hypothesized that the NK cell response to FcR stimulation would be enhanced by the administration of IL-21. Human NK cells cultured with IL-21 and immobilized IgG or human breast cancer cells coated with a therapeutic mAb (trastuzumab) secreted large amounts of IFN-γ. Increased secretion of TNF-α and the chemokines IL-8, MIP-1α, and RANTES was also observed under these conditions. NK cell IFN-γ production was dependent on distinct signals mediated by the IL-21R and the FcR and was abrogated in STAT1-deficient NK cells. Supernatants derived from NK cells that had been stimulated with IL-21 and mAb-coated breast cancer cells were able to drive the migration of naive and activated T cells in an in vitro chemotaxis assay. IL-21 also enhanced NK cell lytic activity against Ab-coated tumor cells. Coadministration of IL-21 and Ab-coated tumor cells to immunocompetent mice led to synergistic production of IFN-γ by NK cells. Furthermore, the administration of IL-21 augmented the effects of an anti-HER2/neu mAb in a murine tumor model, an effect that required IFN-γ. These findings demonstrate that IL-21 significantly enhances the NK cell response to Ab-coated targets and suggest that IL-21 would be an effective adjuvant to administer in combination with therapeutic mAbs.
- Published
- 2006
- Full Text
- View/download PDF
39. Natural Killer Cells Produce T Cell–Recruiting Chemokines in Response to Antibody-Coated Tumor Cells
- Author
-
Julie M. Roda, Robin Parihar, Gerard J. Nuovo, William E. Carson, Susheela Tridandapani, and Cynthia M. Magro
- Subjects
Cancer Research ,Antibodies, Neoplasm ,T-Lymphocytes ,Breast Neoplasms ,Receptors, Fc ,Adenocarcinoma ,CD8-Positive T-Lymphocytes ,Biology ,Antibodies, Monoclonal, Humanized ,Lymphocyte Activation ,CCL5 ,Natural killer cell ,Mice ,Interleukin 21 ,Cell Movement ,Cell Line, Tumor ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,CCL17 ,CXCL14 ,Mice, Inbred BALB C ,Lymphokine-activated killer cell ,Clinical Trials, Phase I as Topic ,Chemotaxis ,Antibodies, Monoclonal ,Trastuzumab ,Natural killer T cell ,Interleukin-12 ,Killer Cells, Natural ,medicine.anatomical_structure ,Oncology ,Immunology ,Interleukin 12 ,Cancer research ,Female ,Chemokines - Abstract
In the current report, we have examined the ability of natural killer (NK) cells to produce T cell–recruiting chemokines following dual stimulation with interleukin (IL)-2 or IL-12 and human breast cancer cells coated with an antitumor antibody (trastuzumab). NK cells stimulated in this manner secreted an array of T cell–recruiting chemotactic factors, including IL-8, macrophage-derived chemokine, macrophage inflammatory protein 1α (MIP-1α), monocyte chemoattractant protein 1, and regulated on activation, normal T-cell expressed and secreted (RANTES), whereas stimulation of NK cells with either agent alone had minimal effect. Furthermore, these factors were functional for T-cell chemotaxis as culture supernatants derived from costimulated NK cells induced migration of both naïve and activated T cells in an in vitro chemotaxis assay. T-cell migration was significantly reduced when neutralizing antibodies to IL-8, MIP-1α, or RANTES were added to culture supernatants before their use in the chemotaxis assay. In addition, coadministration of trastuzumab-coated tumor cells and IL-12 to mice led to enhanced serum MIP-1α. As a clinical correlate, we examined the chemokine content of serum samples from breast cancer patients enrolled on a phase I trial of trastuzumab and IL-12, and found elevated levels of IL-8, RANTES, IFN-γ inducible protein 10, monokine induced by IFN-γ, and MIP-1α, specifically in those patients that experienced a clinical benefit. Sera from these patients exhibited the ability to direct T-cell migration in a chemotaxis assay, and neutralization of chemokines abrogated this effect. These data are the first to show chemokine production by NK cells, specifically in response to stimulation with antibody-coated tumor cells, and suggest a potential role for NK cell–derived chemokines in patients receiving therapeutic monoclonal antibodies. (Cancer Res 2006; 66(1): 517-26)
- Published
- 2006
- Full Text
- View/download PDF
40. Src Homology 2–Containing Inositol 5′-Phosphatase 1 Negatively Regulates IFN-γ Production by Natural Killer Cells Stimulated with Antibody-Coated Tumor Cells and Interleukin-12
- Author
-
Amy K. Ferketich, Rossana Trotta, William E. Carson, Michael A. Caligiuri, Robin Parihar, Susheela Tridandapani, and Julie M. Roda
- Subjects
Cancer Research ,Fc receptor ,Biology ,Transfection ,CD49b ,Natural killer cell ,Interferon-gamma ,Mice ,Interleukin 21 ,medicine ,Animals ,Humans ,IL-2 receptor ,Phosphorylation ,Extracellular Signal-Regulated MAP Kinases ,Mice, Knockout ,Lymphokine-activated killer cell ,Janus kinase 3 ,Inositol Polyphosphate 5-Phosphatases ,Receptors, IgG ,Antibody-Dependent Cell Cytotoxicity ,Receptors, Interleukin-12 ,Receptors, Interleukin ,Interleukin-12 ,Phosphoric Monoester Hydrolases ,Cell biology ,Enzyme Activation ,Killer Cells, Natural ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Oncology ,Biochemistry ,Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases ,Interleukin 12 ,biology.protein ,Female - Abstract
We have previously shown that natural killer (NK) cells secrete a distinct profile of immunomodulatory cytokines in response to dual stimulation with antibody-coated tumor cells and interleukin-12 (IL-12). This NK cell cytokine response is dependent on synergistic signals mediated by the activating receptor for the Fc portion of IgG (FcγRIIIa) and the IL-12 receptor (IL-12R), both constitutively expressed on NK cells. The phosphatase Src homology 2–containing inositol 5′-phosphatase 1 (SHIP1) is known to exert inhibitory effects on Fc receptor (FcR) signaling via its enzymatic activity on phosphatidylinositol 3-kinase (PI3-K) products within many cells of the immune system, most notably mast cells, B cells, and monocytes. However, its activity in the context of FcR activation on NK cells has not been fully explored. The current study focused on the regulation of FcγRIIIa-induced NK cell cytokine production by SHIP1. Inhibitor studies showed that NK cell IFN-γ production following FcR stimulation in the presence of IL-12 depended, in part, on the downstream products of PI3-K. Overexpression of wild-type (WT) SHIP1, but not a catalytic-deficient mutant, via retroviral transfection of primary human NK cells, resulted in a >70% reduction of NK cell IFN-γ production in response to costimulation. In addition, NK cells from SHIP1−/− mice produced 10-fold greater amounts of IFN-γ following culture with antibody-coated tumor cells plus IL-12 compared with NK cells from WT mice. Further, activation of the mitogen-activated protein kinase (MAPK) family member extracellular signal-regulated kinase (Erk; a downstream target of PI3-K) was significantly enhanced within SHIP1−/− NK cells compared with WT NK cells following costimulation. Pharmacologic inhibition of Erk activity, but not Jnk MAPK activity, led to significantly decreased IFN-γ production from both SHIP1−/− and WT NK cells under these conditions. These results are the first to show a physiologic role for SHIP1 in the regulation of NK cell cytokine production and implicate PI3-K in the induction of MAPK signal transduction following costimulation of NK cells via the FcR and the IL-12R.
- Published
- 2005
- Full Text
- View/download PDF
41. CpG-Containing Oligodeoxynucleotides Act through TLR9 to Enhance the NK Cell Cytokine Response to Antibody-Coated Tumor Cells
- Author
-
Robin Parihar, William E. Carson, and Julie M. Roda
- Subjects
Cytotoxicity, Immunologic ,Adoptive cell transfer ,Chemokine ,CpG Oligodeoxynucleotide ,medicine.medical_treatment ,Immunology ,Receptors, Cell Surface ,Antibodies, Monoclonal, Humanized ,Interferon-gamma ,Mice ,Interleukin 21 ,Adjuvants, Immunologic ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Immunology and Allergy ,Cells, Cultured ,Mice, Inbred BALB C ,biology ,Antibodies, Monoclonal ,TLR9 ,hemic and immune systems ,Trastuzumab ,Adoptive Transfer ,Molecular biology ,DNA-Binding Proteins ,Killer Cells, Natural ,Cytokine ,Oligodeoxyribonucleotides ,CpG site ,Immunoglobulin G ,Toll-Like Receptor 9 ,biology.protein ,Interleukin 12 ,Cytokines ,CpG Islands ,Female ,Neoplasm Transplantation - Abstract
Bacterial DNA contains a high frequency of unmethylated CpG motifs that stimulate immune cells via TLR9. NK cells express a low-affinity activating receptor for the Fc portion of IgG (FcγRIIIa), but were not thought to express TLR9 protein. The direct response of NK cells to CpG oligodeoxynucleotides (ODN) in the presence of FcR stimulation was investigated. Human NK cells cultured in the presence of CpG ODN plus immobilized IgG or Ab-coated tumor cells secreted large amounts of IFN-γ (>2000 pg/ml), whereas cells stimulated with Ab alone, CpG ODN alone, or Ab and control ODN produced negligible amounts. Enhanced secretion of IL-8, macrophage-derived chemokine, and MIP-1α was also observed after costimulation. NK cell cytokine production was not the result of interactions with APCs or their cytokine products. Flow cytometric analysis revealed that 36 ± 3.5% of human NK cells expressed basal levels of TLR9. TLR9 expression in human NK cells was confirmed by immunoblot analysis. Only TLR9-expressing NK cells responded to CpG ODN and Ab, because cytokine production was not observed in NK cells from TLR9-deficient mice. Mice receiving CpG ODN and HER2/neu-positive tumor cells treated with an anti-HER2 Ab exhibited enhanced systemic levels of IFN-γ compared with mice receiving either agent alone. TLR9−/− animals reconstituted with TLR9+/+ NK cells secreted IFN-γ in response to CpG ODN and Ab-coated tumor cells. These findings indicate that CpG ODN can directly enhance the NK cell cytokine response to Ab-coated targets via activation of TLR9.
- Published
- 2005
- Full Text
- View/download PDF
42. A Phase I Study of Interleukin 12 with Trastuzumab in Patients with Human Epidermal Growth Factor Receptor-2-Overexpressing Malignancies
- Author
-
Rhonda Jensen, Adrian Lewis, Donn C. Young, Julie Dierksheide, Charles L. Shapiro, Padma Nadella, Cynthia M. Magro, Carrie De Hoff, Robin Parihar, Anne M. VanBuskirk, and William E. Carson
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Interferon-gamma production ,business.industry ,medicine.medical_treatment ,Interleukin ,medicine.disease ,Breast cancer ,Cytokine ,Trastuzumab ,Internal medicine ,Immunology ,Monoclonal ,Interleukin 12 ,medicine ,Interferon gamma ,business ,medicine.drug - Abstract
Purpose: On the basis of preclinical studies, we hypothesized that interleukin (IL)12 would potentiate the antitumor actions of an antihuman epidermal growth factor receptor-2 (HER2) monoclonal antibody (trastuzumab). We conducted a Phase I trial to determine the safety and optimal biological dose of IL-12 when given in combination with trastuzumab.Patients and Methods: Patients with metastatic HER2-positive malignancies received trastuzumab on day 1 of each weekly cycle. Beginning in week 3, patients also received intravenous injections of IL-12 on days 2 and 5. The IL-12 component was dose-escalated within cohorts of 3 patients (30, 100, 300, or 500 ng/kg). Correlative assays were conducted using serum samples and peripheral blood cells obtained during the course of therapy.Results: Fifteen patients were treated, including 12 with HER2 2+ or 3+ breast cancer. The regimen was well tolerated with IL-12-induced grade 1 nausea and grade 2 fatigue predominating. Evaluation of dose-limiting toxicity and biological end points suggested that the 300 ng/kg dose was both the maximally tolerated dose and the optimal biological dose of IL-12 for use in combination with trastuzumab. Two patients with HER2 3+ breast cancer within the 500 ng/kg dose level experienced grade 1 asymptomatic decreases in left ventricular ejection fraction of 12% and 19% after 3 and 10 months of therapy, respectively. There was one complete response in a patient with HER2 3+ breast cancer metastatic to the axillary, mediastinal, and supraclavicular nodes, and 2 patients with stabilization of bone disease lasting 10 months and >12 months, respectively. Correlative assays showed sustained production of interferon (IFN)γ by natural killer cells only in those patients experiencing a clinical response or stabilization of disease. Elevated serum levels of macrophage inflammatory protein-1α, tumor necrosis factor-α, and the antiangiogenic factors IFN-γ inducible protein-10 and monokine induced by γ were also observed in these patients. Patient genotyping suggested that a specific IFN-γ gene polymorphism might have been associated with increased IFN-γ production. The ability of patient peripheral blood cells to conduct antibody-dependent cellular cytotoxicity against tumor targets in vitro did not correlate with clinical response or dose of IL-12.Conclusions: The addition of IL-12 to trastuzumab therapy did not appear to enhance the efficacy of this antibody treatment. Sustained production of IFN-γ and other cytokines were observed in three patients: One who exhibited a complete response and two others who had stabilization of disease lasting over 6 months. Given the small sample size and heterogeneity of the patient population, the effects of IL-12 on the innate immune response to trastuzumab therapy should be further explored in the context of a larger clinical trial.
- Published
- 2004
- Full Text
- View/download PDF
43. Natural killer cells contribute to the lethality of a murine model of Escherichia coli infection
- Author
-
Cynthia M. Magro, Robin Parihar, William E. Carson, Brian D. Badgwell, Julie Dierksheide, and Thomas A. Russo
- Subjects
Umbilical Veins ,CD3 Complex ,medicine.medical_treatment ,Receptors, Antigen, T-Cell ,Mice, Transgenic ,Biology ,Proinflammatory cytokine ,Natural killer cell ,Interferon-gamma ,Mice ,Interleukin 21 ,Immune system ,Antigen ,Sepsis ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,Phosphorylation ,Cyclophosphamide ,Cells, Cultured ,Escherichia coli Infections ,Etoposide ,Mice, Inbred BALB C ,Tumor Necrosis Factor-alpha ,NF-kappa B ,Intercellular Adhesion Molecule-1 ,DNA-Binding Proteins ,Killer Cells, Natural ,Mice, Inbred C57BL ,Survival Rate ,Methotrexate ,STAT1 Transcription Factor ,medicine.anatomical_structure ,Cytokine ,Doxorubicin ,Immunology ,Trans-Activators ,Interleukin 12 ,Female ,Surgery ,Tumor necrosis factor alpha ,Endothelium, Vascular ,Injections, Intraperitoneal ,Spleen ,Interleukin-1 - Abstract
Background. Cooperative interactions between natural killer (NK) cells and macrophages occur normally during the course of the early immune response to bacterial, protozoal, and viral pathogens, with each cellular compartment providing the other with critical stimulatory factors. We conducted the present study to determine whether NK cells contribute to the dysregulated immune response that accompanies septic shock. Methods. An analysis of the lethality of Escherichia coli CP9 was conducted in mice that had been depleted of NK cells via the injection of an anti-asialo GM1 antibody and in CD ϵ transgenic mice that are deficient in both NK cells and T cells. The 2 groups of mice were analyzed for serum levels of interferon-γ, tumor necrosis factor-α, and interleukin-1β as well as activation of NFκB and STAT1, 2 proinflammatory transcription factors. Results. NK cell-depleted and NK cell-deficient mice exhibited 80% survival in the face of an intraperitoneal bacterial challenge, whereas control mice all died within 12 hours. Serum levels of proinflammatory cytokines were markedly reduced in NK-depleted mice. NF κB and STAT1 activation were also reduced. NK-depleted mice exhibited less inflammation within multiple organs on histologic analysis. Conclusions. These results show that NK cells may contribute to the lethality of bacterial infections via effects on cytokine production. (Surgery 2002;132:205-12.)
- Published
- 2002
- Full Text
- View/download PDF
44. Administration of two macrophage-derived interferon-γ-inducing factors (IL-12 and IL-15) induces a lethal systemic inflammatory response in mice that is dependent on natural killer cells but does not require interferon-γ
- Author
-
Robin Parihar, Yan Hu, Jennifer L. Biber, William E. Carson, Michael A. Caligiuri, Saad Jabbour, Page Bouchard, Julie Dierksheide, and Heinz Baumann
- Subjects
Time Factors ,CD3 Complex ,medicine.medical_treatment ,Immunology ,Receptors, Antigen, T-Cell ,Apoptosis ,Mice, Transgenic ,Inflammation ,Mice, SCID ,Biology ,Systemic inflammation ,Receptors, Tumor Necrosis Factor ,Interferon-gamma ,Mice ,medicine ,Animals ,Intestinal Mucosa ,Interleukin-15 ,Mice, Knockout ,Mice, Inbred BALB C ,Lymphokine-activated killer cell ,Dose-Response Relationship, Drug ,Macrophages ,Shock ,STAT4 Transcription Factor ,Natural killer T cell ,Interleukin-12 ,DNA-Binding Proteins ,Killer Cells, Natural ,Mice, Inbred C57BL ,STAT1 Transcription Factor ,Cytokine ,Interleukin 15 ,Toxicity ,Trans-Activators ,Interleukin 12 ,Cytokines ,Female ,medicine.symptom ,Acute-Phase Proteins - Abstract
Activation of macrophages by microbes results in the rapid production of monokines (e.g., interleukin-12 (IL-12), IL-15, and IL-18), which induce production of interferon-gamma (IFN-gamma) by natural killer (NK) cells. We examined the effects of administering IL-15 in combination with IL-12 in a murine toxicity model to determine how these two cytokines might contribute to the inflammatory state that accompanies infectious processes. The daily, simultaneous administration of IL-15 (3 x 10(5)U) and IL-12 (1 microg) to normal mice resulted in shock and 100% mortality within 3-7 days, whereas minimal toxicity was observed following the administration of IL-15 or IL-12 alone. Mice treated with IL-15 plus IL-12 exhibited lesions of the gastrointestinal tract, elevated serum levels of acute phase reactants and pro-inflammatory cytokines, and NK cell apoptosis. Neutralization of IFN-gamma, TNF-alpha, and IL-1beta was not protective in cytokine-treated mice, however, toxicity and death could be completely abrogated by depletion of NK cells. Mice deficient in the STAT4 transcription factor also exhibited complete protection while mice deficient in IFN-gamma or its downstream mediator, STAT1, did not. These findings suggest that cytokine- stimulated NK cells are able to promote systemic inflammation via the induction of STAT4-responsive genes other than IFN-gamma or TNF-alpha.
- Published
- 2002
- Full Text
- View/download PDF
45. Two-pronged Cell Therapy: Engineering NK cells to target CD22 and redirect bystander T cells to CD19 for the adoptive immunotherapy of B-cell malignancies
- Author
-
Arpad Szoor, Mireya Paulina Velasquez, Challice Lee Bonifant, Abishek Vaidya, Lorenzo Brunetti, Michael Gundry, Robin Parihar, Margaret Goodell, and Stephen Gottschalk
- Subjects
Immunology ,Immunology and Allergy - Abstract
CD19-CAR T cells have shown antitumor activity for B-cell derived malignancies, but they also have shown limitations regarding their toxicity and development of CD19- escape variants. Given their short lifespan and potent cytolytic function NK cells are attractive candidate effectors to express CARs. The goal of this project was to devise a strategy to genetically modify NKs that express a CAR to target CD22 and secrete a CD19-specific T-cell engager (CD19-ENG) to redirect T cells to B-cell leukemias. NKs were transduced with a retroviral vector encoding a CD22-CAR and/or CD19-ENG molecules. To mimic immune escape, CD19 or CD22 knockout (ko) leukemia cells (BV173) were generated. We performed coculture and cytotoxicity assays using CD22 and/or CD19+ targets with NKs +/− T cells. In the absence of T cells, CD22-CAR +/− CD19-ENG NK cells recognized tumor cells in an antigen-dependent manner as judged by cytokine production and tumor killing. Moreover, CD22 CAR +/− NK cells secreting engager molecules efficiently redirected T cells in the presence of CD19+ targets. In vivo, CD22-CAR/CD19-ENG NK cells recognized tumor cells in an antigen-dependent manner, redirected T cells to tumor cells, and induced significant regression of leukemia in xenograft models in comparison to mice treated with CD22-CAR NK cells or NK cells targeting irrelevant antigens. We have generated for the first time NK cells that kill B-cell malignancies through a CAR and simultaneously redirect bystander T cells to a 2nd B-cell antigen to enhance antitumor effects and prevent immune escape. Genetic modification of NK cells to enhance their antitumor activity and redirect bystander T cells may represent a promising addition to current cell therapies for B-cell malignancies.
- Published
- 2017
- Full Text
- View/download PDF
46. Monocyte expression of MICA enhances the Natural Killer Cell response to antibody-coated tumor targets
- Author
-
Eric Leudke, Prashant Trikha, Robin Parihar, Amanda Harper, Neela Bhave, William E. Carson, and Kallan S. Opheim
- Subjects
Pharmacology ,Cancer Research ,Lymphokine-activated killer cell ,Janus kinase 3 ,Monocyte ,Immunology ,Autologous Monocytes ,Biology ,NKG2D ,Molecular biology ,Natural killer cell ,Interleukin 21 ,medicine.anatomical_structure ,Oncology ,Poster Presentation ,medicine ,Interleukin 12 ,Molecular Medicine ,Immunology and Allergy - Abstract
Natural Killer (NK) cells are large granular lymphocytes that are uniquely equipped to promote the anti-tumor response via communication with other immune cell types in the tumor microenvironment. We have shown that NK cells secrete a distinct profile of immune stimulatory factors e.g., interferon-gamma (IFN-γ) with potent anti-tumor activity in response to dual stimulation with antibody (Ab)-coated tumor cells and cytokines, such as interleukin (IL)-12 and that this response is significantly enhanced 10 fold in the presence of autologous monocytes (p < 0.05). Similar results were obtained when NK cell secretion of TNF-alpha (a) and MIP1-a were examined. This enhancement of the NK cell cytokine response by monocytes held true for every known stimulator of NK cell IFN-γ production. Further, autologous monocytes significantly increased the ability of IL-12 activated NK cells to lyse Trastuzumab-coated breast cancer cells in an ADCC assay (p < 0.05). Monocyte enhancement of NK cell activity was shown to be dependent on direct cell-cell contact as determined by a transwell assay. We hypothesized that NK cell effector functions against Ab-coated tumor cells were enhanced via binding of the stimulatory ligand MICA on monocytes to the NKG2D receptor on NK cells. Activation of monocytes with bacterial components (e.g., LPS) or IFN-a led to increased surface expression of the NKG2D ligand MICA and further enhanced the ability of monocytes to act as stimulators of NK cell cytokine secretion. The stimulatory effects of MICA-positive monocytes were duplicated via use of a MICA over-expressing cell line (C1R-MICA) (inducing a 4 fold increase in IFN-γ production) but not the parental MICA-negative cell line. Down-modulation of MICA via siRNA or incubation with a MICA neutralizing FAb fragment prior to co-culture of monocytes with NK cells led to a significant reduction in NK cell IFN-γ secretion (p < 0.05). Blockade of NKG2D on NK cells with a neutralizing Ab also reduced IFN-γ production in this co-culture system. In vivo, depletion of monocytes from Balb/c mice resulted in decreased IFN-γ production by murine NK cells upon exposure to Trastuzumab-coated tumor cells and IL-12. Extended depletion of monocytes in vivo abrogated the immune response to combination therapy with Trastuzumab and IL-12, resulting in significantly larger tumors in monocyte-depleted mice in comparison to mock-depleted controls (p < 0.02). These data suggest that direct cell-cell interaction between NK cells and monocytes may constitute an important mechanism for enhancement of the NK cell anti-tumor response in the setting of monoclonal Ab therapy for cancer.
- Published
- 2014
- Full Text
- View/download PDF
47. Interleukin-2 enhances the natural killer cell response to Herceptin-coated Her2 /neu-positive breast cancer cells
- Author
-
José Baselga, Robin Parihar, Neal J. Meropol, Michael A. Caligiuri, Julie Dierksheide, William E. Carson, Matthew J. Lindemann, and Nicola Personeni
- Subjects
Lymphokine-activated killer cell ,Janus kinase 3 ,Immunology ,Biology ,Natural killer T cell ,Molecular biology ,CD49b ,Natural killer cell ,Interleukin 21 ,medicine.anatomical_structure ,Interleukin 12 ,medicine ,Immunology and Allergy ,IL-2 receptor ,skin and connective tissue diseases - Abstract
The Her2 / neu (c-erbB-2) oncogene encodes a 185-kDa protein tyrosine kinase which is overexpressed in 20 % of breast adenocarcinomas and is recognized by a humanized anti-Her2 / neu monoclonal antibody (mAb) (rhu4D5 or Herceptin). Natural killer (NK) cells are capable of mediating antibody-dependent cell cytotoxicity (ADCC) against antibody-coated targets via their expression of a low-affinity receptor for IgG (FcγRIII or CD16). NK cells can be expanded in cancer patients via the administration of low-dose interleukin-2 (IL-2) and become potent cytotoxic effectors following exposure to high doses of IL-2. We tested IL-2-activated NK cells against Her2 / neu+ (MCF-7Her2 / neu) and Her2 / neu– (MDA-468) breast cancer cell lines in a 4-h 51Cr-release cytotoxicity assay in the presence or absence of rhu4D5 mAb (effector : target ratio = 10 : 1). Specific lysis of rhu4D5-coated MCF-7Her2 / neu and MDA-468 target cells by IL-2-activated NK cells was 35 % and 3 %, respectively (p
- Published
- 2001
- Full Text
- View/download PDF
48. Two-Pronged Cell Therapy for B-Cell Malignancies: Engineering NK Cells to Target CD22 and Redirect Bystander T Cells to CD19
- Author
-
Arpad Szoor, Margaret A. Goodell, Stephen Gottschalk, Robin Parihar, Lorenzo Brunetti, Michael C. Gundry, Mireya Paulina Velasquez, Challice L. Bonifant, and Abishek Vaidya
- Subjects
Adoptive cell transfer ,medicine.medical_treatment ,Immunology ,chemical and pharmacologic phenomena ,Biochemistry ,CD19 ,Viral vector ,Cell therapy ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,immune system diseases ,hemic and lymphatic diseases ,medicine ,B cell ,biology ,Chemistry ,hemic and immune systems ,Cell Biology ,Hematology ,medicine.anatomical_structure ,Cytokine ,Interleukin 15 ,Cancer research ,biology.protein ,030215 immunology - Abstract
Background: T-cell therapy with CD19-CAR T cells has been very successful for the treatment of B-cell derived malignancies in humans. However, cytokine release syndrome, neurotoxicity, and development of CD19- escape variants have emerged as potential limitations. Developing CAR NK-cell based therapies might overcome some of these side effects since NK cells do not rapidly expand or persist long-term after adoptive transfer. However, CAR NK-cell therapies are less effective than CAR T-cell therapies in preclinical models. To overcome these limitations we have devised a strategy to genetically modify NK cells with CD22-CARs and CD19/CD3-bispecific T-cell engager (CD19-ENG) molecules. These NK cells should not only kill CD22+ B cells directly, but also redirect bystander T cells to malignant CD19+ B cells, enhancing antitumor effects and preventing immune escape. Methods: NK cells were generated using K562s expressing 41BBL and membrane bound IL15, and genetically modified with a retroviral vector encoding a CD22-CAR with a 41BB.ζ endodomain and/or a retroviral vector encoding CD19-ENG and mOrange separated by an IRES. To mimic immune escape, CD19 or CD22 knockout (ko) Ph+ leukemia cells (BV173) were generated by CRISPR/cas9. The effector function of genetically modified NK cells was evaluated using standard immunological assays. Results: After transduction 70-80% of NK cells expressed CD22-CARs, and ~50% expressed CD22-CARs and CD19-ENGs as judged by FACS analysis. We performed coculture and cytotoxicity assays using non-transduced (NT), CD22-CAR, CD19-ENG, and CD22-CAR/CD19-ENG NK cells as effectors and BV173 (CD19+/CD22+), BV173.koCD19, BV173.koCD22, Daudi (CD19+/CD22+), and KG1a (CD19-,CD22-) as targets. Cocultures were preformed +/- T cells and after 24 hours IFNγ and IL2 was determined by ELISA. In the absence of T cells, CD22-CAR and CD22-CAR/CD19-ENG NK cells only recognized CD22+ targets as judged by IFNγ production. Moreover, CD22-CAR/CD19-ENG and CD19-ENG NK cells efficiently redirected T cells to secrete IFNγ in the presence of CD19+/CD22- targets. No NK-cell population produced IL2, however CD22-CAR/CD19-ENG and CD19-ENG NK cells induced IL2 production of T cells in the presence of CD19+ targets. No significant cytokine production was observed in the absence of antigen (media, KG1a). Specificity of generated NK cells was confirmed in cytotoxicity assays. In vivo studies to confirm our in vitro findings are in progress. Conclusions: We have generated for the first time NK cells that kill B-cell malignancies through a CAR (CD22) and simultaneously redirect bystander T cells to a 2nd B-cell antigen (CD19) to enhance antitumor effects and prevent immune escape. Genetic modification of NK cells to enhance their antitumor activity and redirect bystander T cells may present a promising addition to current cell therapies for B-cell malignancies. Disclosures No relevant conflicts of interest to declare.
- Published
- 2016
- Full Text
- View/download PDF
49. IL-12 enhances the antitumor actions of trastuzumab via NK cell IFN-γ production
- Author
-
Volodymyr Karpa, Robin Parihar, Aruna Mani, David Jarjoura, Natalie B. Jones, Krista LaPerle, Elizabeth Biller, Abhik Ray Chaudhury, Julie M. Roda, Bethany L. Mundy-Bosse, Amy Lehman, Alena Cristina Jaime-Ramirez, Richard W. Burry, Sri Vidya Kondadasula, Tracey L. Papenfuss, and William E. Carson
- Subjects
Receptor, ErbB-2 ,medicine.medical_treatment ,Immunology ,Antineoplastic Agents ,Biology ,Adenocarcinoma ,Antibodies, Monoclonal, Humanized ,Article ,Interleukin 21 ,Interferon-gamma ,Mice ,Random Allocation ,Immune system ,medicine ,Immunology and Allergy ,Animals ,Interferon gamma ,Mice, Inbred BALB C ,Antibodies, Monoclonal ,Trastuzumab ,Cytotoxicity Tests, Immunologic ,Interleukin-12 ,Up-Regulation ,Monokine ,Killer Cells, Natural ,Cytokine ,Colonic Neoplasms ,Cancer research ,Interleukin 12 ,Tumor necrosis factor alpha ,Female ,CD8 ,medicine.drug - Abstract
The antitumor effects of therapeutic mAbs may depend on immune effector cells that express FcRs for IgG. IL-12 is a cytokine that stimulates IFN-γ production from NK cells and T cells. We hypothesized that coadministration of IL-12 with a murine anti-HER2/neu mAb (4D5) would enhance the FcR-dependent immune mechanisms that contribute to its antitumor activity. Thrice-weekly therapy with IL-12 (1 μg) and 4D5 (1 mg/kg) significantly suppressed the growth of a murine colon adenocarcinoma that was engineered to express human HER2 (CT-26HER2/neu) in BALB/c mice compared with the result of therapy with IL-12, 4D5, or PBS alone. Combination therapy was associated with increased circulating levels of IFN-γ, monokine induced by IFN-γ, and RANTES. Experiments with IFN-γ–deficient mice demonstrated that this cytokine was necessary for the observed antitumor effects of therapy with IL-12 plus 4D5. Immune cell depletion experiments showed that NK cells (but not CD4+ or CD8+ T cells) mediated the antitumor effects of this treatment combination. Therapy of HER2/neu-positive tumors with trastuzumab plus IL-12 induced tumor necrosis but did not affect tumor proliferation, apoptosis, vascularity, or lymphocyte infiltration. In vitro experiments with CT-26HER2/neu tumor cells revealed that IFN-γ induced an intracellular signal but did not inhibit cellular proliferation or induce apoptosis. Taken together, these data suggest that tumor regression in response to trastuzumab plus IL-12 is mediated through NK cell IFN-γ production and provide a rationale for the coadministration of NK cell-activating cytokines with therapeutic mAbs.
- Published
- 2011
50. Abstract PR005: TGF-beta1 primed myeloid derived suppressor cells decrease tumor growth and lose their ability to inhibit T cell proliferation via iNOS downregulation
- Author
-
Rosemarie Krupar, Indu Varier, Padmini Jayaraman, Robin Parihar, Andrew G. Sikora, and Falguni Parikh
- Subjects
Cancer Research ,Tumor microenvironment ,Myeloid ,medicine.diagnostic_test ,Angiogenesis ,medicine.medical_treatment ,T cell ,Immunology ,CD28 ,Biology ,Flow cytometry ,medicine.anatomical_structure ,Cytokine ,Cancer research ,medicine ,Myeloid-derived Suppressor Cell - Abstract
Background: Myeloid derived suppressor cells or MDSC are a heterogeneous population of bone marrow-derived cells that consist of myeloid progenitor and immature. Cancer-induced pro-inflammatory signals recruit MDSC from the bone marrow and maintain them in an undifferentiated state. MDSCs inhibit T cell proliferation via up regulation of iNOS and ROS thereby suppressing antitumor immune responses. MDSC have also been shown to promote tumor growth by stimulating angiogenesis and other mechanisms. TGF-b1 is a pleiotropic cytokine abundantly expressed in the tumor microenvironment with diverse effects on myeloid, lymphoid and tumor cells. The aim of this study is to determine the effect of TGF-b1 in the generation and function of MDSC, including its effects on T cell proliferation and tumor growth. Methods: Ex vivo MDSC generation: Bone marrow progenitor cells were derived from WT C57bl/6 mice and co-cultured with MTEC (mouse tonsil epithelial cells transformed with HPV16 E6+E7 oncogenes and H-Ras ) tumor supernatants in the presence or absence of TGF-β1 for 5 days at 370C. Cells were then harvested, processed into single cell suspensions, and stained for MDSC surface markers, DAF-DA (to determine nitric oxide levels),iNOS and other functional markers analysis was performed by flow cytometry (FACS). MDSC functional assay: A) T cell proliferation assay MDSCs were generated in the presence or absence of TGF-β1 with supernatants from MTEC cells and co-cultured with CFSE labeled T cells activated with anti CD3 and anti CD28 antibodies. T cell proliferation was measured by using CFSE dilution, which was analyzed by flow cytometry. B) Effect of MDSC on tumor growth Control and TGF-β1 conditioned MDSC were co-cultured with MTEC tumor (grown as spheroids) for 72 hrs at the end of which histological sections of the spheroids were prepared and analyzed for tumor proliferation by Ki-67 staining. Sorted MDSCs were also co-cultured with T-hep3 cells grown in a monolayer (human oral cancer line) and tumor growth was determined by flow cytometry. Results: While control MDSC suppressed T cell proliferation in a dose-dependent fashion, we observed that TGF-β1 primed MDSCs lost the ability to inhibit T cell proliferation. Further, TGF-β1 primed MDSC inhibited tumor growth in an ex vivo co-culture system. Histological sections of tumor spheroid / MDSC co-cultures revealed diminshed ki-67 expression in spheroids cultured with TGF-B1 conditioned MDSC compared to control. Upon further examining the cellular mechanism, it was seen that TGF-β1 treated MDSCs down regulate iNOS expression and produced decreased amounts of nitric oxide compared to their control counterparts, without altering the expression of other MDSC functional markers like arginase, PD-1 and PD-L1. Conclusions: We conclude that TGF-β1 reprograms MDSC via an iNOS/NO dependent mechanism to a) T cell suppressive capacity and b) inhibit tumor cell growth. These observations have a direct translational implication wherein the inherent pro-tumor nature of MDSCs could potentially be reprogrammed with TGF-β1 and directed toward the tumor thereby suppressing tumor growth. Citation Format: Padmini Jayaraman, Falguni Parikh, Rosemarie Krupar, Robin Parihar, Indu Varier, Andrew Sikora. TGF-beta1 primed myeloid derived suppressor cells decrease tumor growth and lose their ability to inhibit T cell proliferation via iNOS downregulation. [abstract]. In: Proceedings of the CRI-CIMT-EATI-AACR Inaugural International Cancer Immunotherapy Conference: Translating Science into Survival; September 16-19, 2015; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(1 Suppl):Abstract nr PR005.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.