37 results on '"Parisa, Malekzadeh"'
Search Results
2. 152 Adoptive T cell therapy targeting somatic p53 mutations
- Author
-
Nikolaos Zacharakis, Parisa Malekzadeh, Peter Kim, Nolan Vale, and Elizabeth Hedges
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2020
- Full Text
- View/download PDF
3. 333 Targeting the apical intracellular checkpoint CISH unleashes T cell neoantigen reactivity and effector program
- Author
-
Matthew Johnson, Ying Hu, Nicholas Restifo, Jared Gartner, Douglas Palmer, Shashank Patel, Maria Parkhurst, Todd Prickett, Parisa Malekzadeh, David McKenna, Zulmarie Franco, Suman Vodnala, Beau Webber, Yogin Patel, Christine Kariya, Walker Lahr, Rigel Kishton, Devikala Gurusamy, Miechaleen Diers, Natalie Wolf, Nicholas Slipek, Darin Sumstad, Lydia Viney, Tom Henley, Tilmann Bürckstümmer, Oliver Baker, Chunhua Yan, Daoud Meerzaman, Kartik Padhan, Winnie Lo, Li Jia, Drew Deniger, R Scott McIvor, Modassir Choudhry, and Branden Moriarity
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2020
- Full Text
- View/download PDF
4. Adoptive Cellular Therapy with Autologous Tumor-Infiltrating Lymphocytes and T-cell Receptor–Engineered T Cells Targeting Common p53 Neoantigens in Human Solid Tumors
- Author
-
Sanghyun P. Kim, Nolan R. Vale, Nikolaos Zacharakis, Sri Krishna, Zhiya Yu, Billel Gasmi, Jared J. Gartner, Sivasish Sindiri, Parisa Malekzadeh, Drew C. Deniger, Frank J. Lowery, Maria R. Parkhurst, Lien T. Ngo, Satyajit Ray, Yong F. Li, Victoria Hill, Maria Florentin, Robert V. Masi, Biman C. Paria, Noam Levin, Alakesh Bera, Elizabeth A. Hedges, Agnes Choi, Praveen D. Chatani, Anup Y. Parikh, Shoshana Levi, Samantha Seitter, Yong-Chen Lu, Zhili Zheng, Todd D. Prickett, Li Jia, Jonathan M. Hernandez, Chuong D. Hoang, Paul F. Robbins, Stephanie L. Goff, Richard M. Sherry, James C. Yang, and Steven A. Rosenberg
- Subjects
Genes, T-Cell Receptor ,Cancer Research ,Lymphocytes, Tumor-Infiltrating ,Antigens, Neoplasm ,Neoplasms ,T-Lymphocytes ,Immunology ,Hematopoietic Stem Cell Transplantation ,Receptors, Antigen, T-Cell ,Humans ,Tumor Suppressor Protein p53 - Abstract
Adoptive cellular therapy (ACT) targeting neoantigens can achieve durable clinical responses in patients with cancer. Most neoantigens arise from patient-specific mutations, requiring highly individualized treatments. To broaden the applicability of ACT targeting neoantigens, we focused on TP53 mutations commonly shared across different cancer types. We performed whole-exome sequencing on 163 patients with metastatic solid cancers, identified 78 who had TP53 missense mutations, and through immunologic screening, identified 21 unique T-cell reactivities. Here, we report a library of 39 T-cell receptors (TCR) targeting TP53 mutations shared among 7.3% of patients with solid tumors. These TCRs recognized tumor cells in a TP53 mutation- and human leucocyte antigen (HLA)-specific manner in vitro and in vivo. Twelve patients with chemorefractory epithelial cancers were treated with ex vivo–expanded autologous tumor-infiltrating lymphocytes (TIL) that were naturally reactive against TP53 mutations. However, limited clinical responses (2 partial responses among 12 patients) were seen. These infusions contained low frequencies of mutant p53–reactive TILs that had exhausted phenotypes and showed poor persistence. We also treated one patient who had chemorefractory breast cancer with ACT comprising autologous peripheral blood lymphocytes transduced with an allogeneic HLA-A*02–restricted TCR specific for p53R175H. The infused cells exhibited an improved immunophenotype and prolonged persistence compared with TIL ACT and the patient experienced an objective tumor regression (-55%) that lasted 6 months. Collectively, these proof-of-concept data suggest that the library of TCRs targeting shared p53 neoantigens should be further evaluated for the treatment of patients with advanced human cancers. See related Spotlight by Klebanoff, p. 919
- Published
- 2022
- Full Text
- View/download PDF
5. Supplementary Figure 5 from Immunologic Recognition of a Shared p53 Mutated Neoantigen in a Patient with Metastatic Colorectal Cancer
- Author
-
Steven A. Rosenberg, David S. Schrump, Isaac Kriley, Biman C. Paria, Scott Kivitz, Satyajit Ray, Todd Prickett, Thomas E. Shelton, Parisa Malekzadeh, Drew Deniger, Anna Pasetto, Jared J. Gartner, Li Jia, Yong-Chen Lu, Eric Tran, Paul F. Robbins, Maria Parkhurst, and Winifred Lo
- Abstract
Supplementary Figure 5
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Tables 1 and 2 from Immunologic Recognition of a Shared p53 Mutated Neoantigen in a Patient with Metastatic Colorectal Cancer
- Author
-
Steven A. Rosenberg, David S. Schrump, Isaac Kriley, Biman C. Paria, Scott Kivitz, Satyajit Ray, Todd Prickett, Thomas E. Shelton, Parisa Malekzadeh, Drew Deniger, Anna Pasetto, Jared J. Gartner, Li Jia, Yong-Chen Lu, Eric Tran, Paul F. Robbins, Maria Parkhurst, and Winifred Lo
- Abstract
Supplementary Tables 1 and 2
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Table 3 from Immunologic Recognition of a Shared p53 Mutated Neoantigen in a Patient with Metastatic Colorectal Cancer
- Author
-
Steven A. Rosenberg, David S. Schrump, Isaac Kriley, Biman C. Paria, Scott Kivitz, Satyajit Ray, Todd Prickett, Thomas E. Shelton, Parisa Malekzadeh, Drew Deniger, Anna Pasetto, Jared J. Gartner, Li Jia, Yong-Chen Lu, Eric Tran, Paul F. Robbins, Maria Parkhurst, and Winifred Lo
- Abstract
Supplementary Table 3
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Figure from Adoptive Cellular Therapy with Autologous Tumor-Infiltrating Lymphocytes and T-cell Receptor–Engineered T Cells Targeting Common p53 Neoantigens in Human Solid Tumors
- Author
-
Steven A. Rosenberg, James C. Yang, Richard M. Sherry, Stephanie L. Goff, Paul F. Robbins, Chuong D. Hoang, Jonathan M. Hernandez, Li Jia, Todd D. Prickett, Zhili Zheng, Yong-Chen Lu, Samantha Seitter, Shoshana Levi, Anup Y. Parikh, Praveen D. Chatani, Agnes Choi, Elizabeth A. Hedges, Alakesh Bera, Noam Levin, Biman C. Paria, Robert V. Masi, Maria Florentin, Victoria Hill, Yong F. Li, Satyajit Ray, Lien T. Ngo, Maria R. Parkhurst, Frank J. Lowery, Drew C. Deniger, Parisa Malekzadeh, Sivasish Sindiri, Jared J. Gartner, Billel Gasmi, Zhiya Yu, Sri Krishna, Nikolaos Zacharakis, Nolan R. Vale, and Sanghyun P. Kim
- Abstract
Supplementary Figure from Adoptive Cellular Therapy with Autologous Tumor-Infiltrating Lymphocytes and T-cell Receptor–Engineered T Cells Targeting Common p53 Neoantigens in Human Solid Tumors
- Published
- 2023
- Full Text
- View/download PDF
9. Data from Immunologic Recognition of a Shared p53 Mutated Neoantigen in a Patient with Metastatic Colorectal Cancer
- Author
-
Steven A. Rosenberg, David S. Schrump, Isaac Kriley, Biman C. Paria, Scott Kivitz, Satyajit Ray, Todd Prickett, Thomas E. Shelton, Parisa Malekzadeh, Drew Deniger, Anna Pasetto, Jared J. Gartner, Li Jia, Yong-Chen Lu, Eric Tran, Paul F. Robbins, Maria Parkhurst, and Winifred Lo
- Abstract
Adoptive cell therapy (ACT) with T cells targeting neoantigens can mediate durable responses in patients with metastatic cancer. Cell therapies targeting common shared antigens for epithelial cancers are not yet broadly available. Here, we report the identification and characterization in one patient of T-cell receptors (TCRs) recognizing mutated p53 p.R175H, which is shared among a subset of patients with cancer. Tumor-infiltrating lymphocytes were screened for recognition of mutated neoantigens in a patient with metastatic colorectal cancer. HLA-A*0201–restricted recognition of mutated p53 p.R175H was identified, and the minimal peptide epitope was HMTEVVRHC. Reactive T cells were isolated by tetramer sorting, and three TCRs were identified. These TCRs mediated recognition of commercially available ovarian cancer, uterine carcinoma, and myeloma cell lines, as well as an NIH patient–derived esophageal adenocarcinoma line that endogenously expressed p53 p.R175H and HLA-A*0201. They also mediated recognition of p53 p.R175H+ colon, breast, and leukemia cell lines after transduction with a retrovirus encoding HLA-A*0201. This work demonstrates that common shared mutated epitopes such as those found in p53 can elicit immunogenic responses and that the application of ACT may be extended to patients with any cancer histology that expresses both HLA-A*0201 and the p53 p.R175H mutation.
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Figures from Immunologic Recognition of a Shared p53 Mutated Neoantigen in a Patient with Metastatic Colorectal Cancer
- Author
-
Steven A. Rosenberg, David S. Schrump, Isaac Kriley, Biman C. Paria, Scott Kivitz, Satyajit Ray, Todd Prickett, Thomas E. Shelton, Parisa Malekzadeh, Drew Deniger, Anna Pasetto, Jared J. Gartner, Li Jia, Yong-Chen Lu, Eric Tran, Paul F. Robbins, Maria Parkhurst, and Winifred Lo
- Abstract
Supplementary Figures 1, 2, 3, 4, 6 and 7
- Published
- 2023
- Full Text
- View/download PDF
11. Data from Adoptive Cellular Therapy with Autologous Tumor-Infiltrating Lymphocytes and T-cell Receptor–Engineered T Cells Targeting Common p53 Neoantigens in Human Solid Tumors
- Author
-
Steven A. Rosenberg, James C. Yang, Richard M. Sherry, Stephanie L. Goff, Paul F. Robbins, Chuong D. Hoang, Jonathan M. Hernandez, Li Jia, Todd D. Prickett, Zhili Zheng, Yong-Chen Lu, Samantha Seitter, Shoshana Levi, Anup Y. Parikh, Praveen D. Chatani, Agnes Choi, Elizabeth A. Hedges, Alakesh Bera, Noam Levin, Biman C. Paria, Robert V. Masi, Maria Florentin, Victoria Hill, Yong F. Li, Satyajit Ray, Lien T. Ngo, Maria R. Parkhurst, Frank J. Lowery, Drew C. Deniger, Parisa Malekzadeh, Sivasish Sindiri, Jared J. Gartner, Billel Gasmi, Zhiya Yu, Sri Krishna, Nikolaos Zacharakis, Nolan R. Vale, and Sanghyun P. Kim
- Abstract
Adoptive cellular therapy (ACT) targeting neoantigens can achieve durable clinical responses in patients with cancer. Most neoantigens arise from patient-specific mutations, requiring highly individualized treatments. To broaden the applicability of ACT targeting neoantigens, we focused on TP53 mutations commonly shared across different cancer types. We performed whole-exome sequencing on 163 patients with metastatic solid cancers, identified 78 who had TP53 missense mutations, and through immunologic screening, identified 21 unique T-cell reactivities. Here, we report a library of 39 T-cell receptors (TCR) targeting TP53 mutations shared among 7.3% of patients with solid tumors. These TCRs recognized tumor cells in a TP53 mutation- and human leucocyte antigen (HLA)-specific manner in vitro and in vivo. Twelve patients with chemorefractory epithelial cancers were treated with ex vivo–expanded autologous tumor-infiltrating lymphocytes (TIL) that were naturally reactive against TP53 mutations. However, limited clinical responses (2 partial responses among 12 patients) were seen. These infusions contained low frequencies of mutant p53–reactive TILs that had exhausted phenotypes and showed poor persistence. We also treated one patient who had chemorefractory breast cancer with ACT comprising autologous peripheral blood lymphocytes transduced with an allogeneic HLA-A*02–restricted TCR specific for p53R175H. The infused cells exhibited an improved immunophenotype and prolonged persistence compared with TIL ACT and the patient experienced an objective tumor regression (-55%) that lasted 6 months. Collectively, these proof-of-concept data suggest that the library of TCRs targeting shared p53 neoantigens should be further evaluated for the treatment of patients with advanced human cancers.See related Spotlight by Klebanoff, p. 919
- Published
- 2023
- Full Text
- View/download PDF
12. Data from Unique Neoantigens Arise from Somatic Mutations in Patients with Gastrointestinal Cancers
- Author
-
Steven A. Rosenberg, Robert P.T. Somerville, John R. Wunderlich, Mojgan Ahmadzadeh, Parisa Malekzadeh, Drew C. Deniger, Gal Cafri, Winifred Lo, Stephanie L. Goff, Frank J. Lowery, Rami Yossef, Anna Pasetto, Scott Kivitz, Lien T. Ngo, Satyajit Ray, Eric Groh, Abraham Sachs, Jessica S. Crystal, Almin Lalani, Mona El-Gamil, Yong F. Li, Gabriel Ivey, Li Jia, Jared J. Gartner, Todd D. Prickett, Eric Tran, Paul F. Robbins, and Maria R. Parkhurst
- Abstract
Immunotherapies can mediate regression of human tumors with high mutation rates, but responses are rarely observed in patients with common epithelial cancers. This raises the question of whether patients with these common cancers harbor T lymphocytes that recognize mutant proteins expressed by autologous tumors that may represent ideal targets for immunotherapy. Using high-throughput immunologic screening of mutant gene products identified via whole-exome sequencing, we identified neoantigen-reactive tumor-infiltrating lymphocytes (TIL) from 62 of 75 (83%) patients with common gastrointestinal cancers. In total, 124 neoantigen-reactive TIL populations were identified, and all but one of the neoantigenic determinants were unique. The results of in vitro T-cell recognition assays demonstrated that 1.6% of the gene products encoded by somatic nonsynonymous mutations were immunogenic. These findings demonstrate that the majority of common epithelial cancers elicit immune recognition and open possibilities for cell-based immunotherapies for patients bearing these cancers.Significance:TILs cultured from 62 of 75 (83%) patients with gastrointestinal cancers recognized neoantigens encoded by 1.6% of somatic mutations expressed by autologous tumor cells, and 99% of the neoantigenic determinants appeared to be unique and not shared between patients.This article is highlighted in the In This Issue feature, p. 983
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Figures from Unique Neoantigens Arise from Somatic Mutations in Patients with Gastrointestinal Cancers
- Author
-
Steven A. Rosenberg, Robert P.T. Somerville, John R. Wunderlich, Mojgan Ahmadzadeh, Parisa Malekzadeh, Drew C. Deniger, Gal Cafri, Winifred Lo, Stephanie L. Goff, Frank J. Lowery, Rami Yossef, Anna Pasetto, Scott Kivitz, Lien T. Ngo, Satyajit Ray, Eric Groh, Abraham Sachs, Jessica S. Crystal, Almin Lalani, Mona El-Gamil, Yong F. Li, Gabriel Ivey, Li Jia, Jared J. Gartner, Todd D. Prickett, Eric Tran, Paul F. Robbins, and Maria R. Parkhurst
- Abstract
Supplementary Figures 1-9
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Data from Unique Neoantigens Arise from Somatic Mutations in Patients with Gastrointestinal Cancers
- Author
-
Steven A. Rosenberg, Robert P.T. Somerville, John R. Wunderlich, Mojgan Ahmadzadeh, Parisa Malekzadeh, Drew C. Deniger, Gal Cafri, Winifred Lo, Stephanie L. Goff, Frank J. Lowery, Rami Yossef, Anna Pasetto, Scott Kivitz, Lien T. Ngo, Satyajit Ray, Eric Groh, Abraham Sachs, Jessica S. Crystal, Almin Lalani, Mona El-Gamil, Yong F. Li, Gabriel Ivey, Li Jia, Jared J. Gartner, Todd D. Prickett, Eric Tran, Paul F. Robbins, and Maria R. Parkhurst
- Abstract
Supplementary Materials and Methods
- Published
- 2023
- Full Text
- View/download PDF
15. Table S1 and S2 from T-cell Responses to TP53 'Hotspot' Mutations and Unique Neoantigens Expressed by Human Ovarian Cancers
- Author
-
Steven A. Rosenberg, Robert P.T. Somerville, David N. Danforth, John R. Wunderlich, Michelle M. Langhan, Rami Yossef, Li Jia, Parisa Malekzadeh, Biman C. Paria, Todd D. Prickett, Jared J. Gartner, Paul F. Robbins, Anna Pasetto, and Drew C. Deniger
- Abstract
Clinical details (S1) and neoepitope details (S2).
- Published
- 2023
- Full Text
- View/download PDF
16. Data from T-cell Responses to TP53 'Hotspot' Mutations and Unique Neoantigens Expressed by Human Ovarian Cancers
- Author
-
Steven A. Rosenberg, Robert P.T. Somerville, David N. Danforth, John R. Wunderlich, Michelle M. Langhan, Rami Yossef, Li Jia, Parisa Malekzadeh, Biman C. Paria, Todd D. Prickett, Jared J. Gartner, Paul F. Robbins, Anna Pasetto, and Drew C. Deniger
- Abstract
Purpose: This was a study prospectively evaluating intratumoral T-cell responses to autologous somatic mutated neoepitopes expressed by human metastatic ovarian cancers.Patients and Methods: Tumor-infiltrating lymphocytes (TIL) were expanded from resected ovarian cancer metastases, which were analyzed by whole-exome and transcriptome sequencing to identify autologous somatic mutations. All mutated neoepitopes, independent of prediction algorithms, were expressed in autologous antigen-presenting cells and then cocultured with TIL fragment cultures. Secretion of IFNγ or upregulation of 41BB indicated a T-cell response.Results: Seven women with metastatic ovarian cancer were evaluated, and 5 patients had clear, dominant T-cell responses to mutated neoantigens, which were corroborated by comparison with the wild-type sequence, identification of the minimal epitope, human leukocyte antigen (HLA) restriction element(s), and neoantigen-specific T-cell receptor(s). Mutated neoantigens were restricted by HLA-B, -C, -DP, -DQ, and/or -DR alleles and appeared to principally arise from random, somatic mutations unique to each patient. We established that TP53 “hotspot” mutations (c.659A>G; p.Y220C and c.733G>A; p.G245S) expressed by two different patients' tumors were both immunogenic in the context of HLA-DRB3*02:02.Conclusions: Mutation-reactive T cells infiltrated ovarian cancer metastases at sufficient frequencies to warrant their investigation as adoptive cell therapy. In addition, transfer of TP53 “hotspot” mutation-reactive T-cell receptors into peripheral blood T cells could be evaluated as a gene therapy for a diverse range of tumor histologies. Clin Cancer Res; 24(22); 5562–73. ©2018 AACR.See related commentary by McNeish, p. 5493
- Published
- 2023
- Full Text
- View/download PDF
17. Materials and Methods and Figures S1-S6 from T-cell Responses to TP53 'Hotspot' Mutations and Unique Neoantigens Expressed by Human Ovarian Cancers
- Author
-
Steven A. Rosenberg, Robert P.T. Somerville, David N. Danforth, John R. Wunderlich, Michelle M. Langhan, Rami Yossef, Li Jia, Parisa Malekzadeh, Biman C. Paria, Todd D. Prickett, Jared J. Gartner, Paul F. Robbins, Anna Pasetto, and Drew C. Deniger
- Abstract
Additional materials and methods. Figure S1. CD4+ T cells responded to USP9X-Y2009C minimal epitope. Figure S2. CD4+ T cells recognized INPP5K-L272V neoantigen. Figure S3. T cells transduced with TCRs recognize Histone H1.5-A71D neoantigen. Figure S4. CD4+ T cells responded to RAPTOR-D654G minimal neoepitope. Figure S5. The p53-G245S minimal neoepitope was mapped to the N-terminus of the 25 amino acid neoantigen. Figure S6. Peptide parsing from Patient 7 demonstrated CTAGE5-E576V, p53-Y220C and HUWE1-F4353S responses.
- Published
- 2023
- Full Text
- View/download PDF
18. Data from Antigen Experienced T Cells from Peripheral Blood Recognize p53 Neoantigens
- Author
-
Drew C. Deniger, Steven A. Rosenberg, Robert P.T. Somerville, Nicholas P. Restifo, Zhiya Yu, Mark Raffeld, Liqiang Xi, Satyajit Ray, Paul F. Robbins, Maria R. Parkhurst, Scott Kivitz, Sanghyun P. Kim, Amy R. Copeland, Abraham Sachs, Meghan L. Good, Mohammad Jafferji, Frank J. Lowery, Biman C. Paria, Gal Cafri, Rami Yossef, and Parisa Malekzadeh
- Abstract
Purpose:The purpose of this study was to evaluate antigen experienced T cells in peripheral blood lymphocytes (PBL) for responses to p53 neoantigens.Experimental Design:PBLs from patients with a mutated TP53 tumor were sorted for antigen-experienced T cells and in vitro stimulation (IVS) was performed with p53 neoantigens. The IVS cultures were stimulated with antigen-presenting cells expressing p53 neoantigens, enriched for 41BB/OX40 and grown with rapid expansion protocol.Results:T-cell responses were not observed in the PBLs of 4 patients who did not have tumor-infiltrating lymphocyte (TIL) responses to mutated TP53. In contrast, 5 patients with TIL responses to mutated TP53 also had similar T-cell responses in their PBLs, indicating that the PBLs and TILs were congruent in p53 neoantigen reactivity. CD4+ and CD8+ T cells were specific for p53R175H, p53Y220C, or p53R248W neoantigens, including a 78% reactive T-cell culture against p53R175H and HLA-A*02:01. Tracking TCRB clonotypes (clonality, top ranked, and TP53 mutation-specific) supported the enrichment of p53 neoantigen–reactive T cells from PBLs. The same T-cell receptor (TCR) from the TIL was found in the IVS cultures in three cases and multiple unique TCRs were found in another patient. TP53 mutation–specific T cells also recognized tumor cell lines bearing the appropriate human leukocyte antigen restriction element and TP53 mutation, indicating these T cells could recognize processed and presented p53 neoantigens.Conclusions:PBL was a noninvasive source of T cells targeting TP53 mutations for cell therapy and can provide a window into intratumoral p53 neoantigen immune responses.See related commentary by Olivera et al., p. 1203
- Published
- 2023
- Full Text
- View/download PDF
19. Supplementary Data from Antigen Experienced T Cells from Peripheral Blood Recognize p53 Neoantigens
- Author
-
Drew C. Deniger, Steven A. Rosenberg, Robert P.T. Somerville, Nicholas P. Restifo, Zhiya Yu, Mark Raffeld, Liqiang Xi, Satyajit Ray, Paul F. Robbins, Maria R. Parkhurst, Scott Kivitz, Sanghyun P. Kim, Amy R. Copeland, Abraham Sachs, Meghan L. Good, Mohammad Jafferji, Frank J. Lowery, Biman C. Paria, Gal Cafri, Rami Yossef, and Parisa Malekzadeh
- Abstract
Table 1: Antibodies used in the study. Table 2: Normalized initial screening data from PBL with a T cell response to mutated TP53. Table 3: TCRB tracking of p53 neoantigen-reactive cells before and after IVS, co-culture, 41BB/OX40 sorting and REP.
- Published
- 2023
- Full Text
- View/download PDF
20. Antigen Experienced T Cells from Peripheral Blood Recognize p53 Neoantigens
- Author
-
Scott Kivitz, Gal Cafri, Drew C. Deniger, Steven A. Rosenberg, Parisa Malekzadeh, Paul F. Robbins, Biman C. Paria, Rami Yossef, Mark Raffeld, Nicholas P. Restifo, Amy R. Copeland, Satyajit Ray, Maria R. Parkhurst, Robert Somerville, Sang-hyun Kim, Meghan L. Good, Zhiya Yu, Abraham Sachs, Liqiang Xi, Frank J. Lowery, and Mohammad S. Jafferji
- Subjects
CD4-Positive T-Lymphocytes ,0301 basic medicine ,Cancer Research ,Lymphocyte ,Receptors, Antigen, T-Cell ,Human leukocyte antigen ,CD8-Positive T-Lymphocytes ,Biology ,Article ,Cell therapy ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,Immune system ,Antigen ,Antigens, Neoplasm ,medicine ,Humans ,Receptor ,integumentary system ,T-cell receptor ,Oncogenes ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Immunology ,Immunotherapy ,Tumor Suppressor Protein p53 ,CD8 - Abstract
Purpose: The purpose of this study was to evaluate antigen experienced T cells in peripheral blood lymphocytes (PBL) for responses to p53 neoantigens. Experimental Design: PBLs from patients with a mutated TP53 tumor were sorted for antigen-experienced T cells and in vitro stimulation (IVS) was performed with p53 neoantigens. The IVS cultures were stimulated with antigen-presenting cells expressing p53 neoantigens, enriched for 41BB/OX40 and grown with rapid expansion protocol. Results: T-cell responses were not observed in the PBLs of 4 patients who did not have tumor-infiltrating lymphocyte (TIL) responses to mutated TP53. In contrast, 5 patients with TIL responses to mutated TP53 also had similar T-cell responses in their PBLs, indicating that the PBLs and TILs were congruent in p53 neoantigen reactivity. CD4+ and CD8+ T cells were specific for p53R175H, p53Y220C, or p53R248W neoantigens, including a 78% reactive T-cell culture against p53R175H and HLA-A*02:01. Tracking TCRB clonotypes (clonality, top ranked, and TP53 mutation-specific) supported the enrichment of p53 neoantigen–reactive T cells from PBLs. The same T-cell receptor (TCR) from the TIL was found in the IVS cultures in three cases and multiple unique TCRs were found in another patient. TP53 mutation–specific T cells also recognized tumor cell lines bearing the appropriate human leukocyte antigen restriction element and TP53 mutation, indicating these T cells could recognize processed and presented p53 neoantigens. Conclusions: PBL was a noninvasive source of T cells targeting TP53 mutations for cell therapy and can provide a window into intratumoral p53 neoantigen immune responses. See related commentary by Olivera et al., p. 1203
- Published
- 2020
- Full Text
- View/download PDF
21. Cancer genes disfavoring T cell immunity identified via integrated systems approach
- Author
-
Rigel J. Kishton, Shashank J. Patel, Amy E. Decker, Suman K. Vodnala, Maggie Cam, Tori N. Yamamoto, Yogin Patel, Madhusudhanan Sukumar, Zhiya Yu, Michelle Ji, Amanda N. Henning, Devikala Gurusamy, Douglas C. Palmer, Roxana A. Stefanescu, Andrew T. Girvin, Winifred Lo, Anna Pasetto, Parisa Malekzadeh, Drew C. Deniger, Kris C. Wood, Neville E. Sanjana, and Nicholas P. Restifo
- Subjects
Antigen Presentation ,Systems Analysis ,Neoplasms ,T-Lymphocytes ,Humans ,Oncogenes ,CRISPR-Cas Systems ,General Biochemistry, Genetics and Molecular Biology - Abstract
Adoptive T cell therapies (ACT) have been curative for a limited number of cancer patients. The sensitization of cancer cells to T cell killing may expand the benefit of these therapies for more patients. To this end, we use a three-step approach to identify cancer genes that disfavor T cell immunity. First, we profile gene transcripts upregulated by cancer under selection pressure from T cell killing. Second, we identify potential tumor gene targets and pathways that disfavor T cell killing using signaling pathway activation libraries and genome-wide loss-of-function CRISPR-Cas9 screens. Finally, we implement pharmacological perturbation screens to validate these targets and identify BIRC2, ITGAV, DNPEP, BCL2, and ERRα as potential ACT-drug combination candidates. Here, we establish that BIRC2 limits antigen presentation and T cell recognition of tumor cells by suppressing IRF1 activity and provide evidence that BIRC2 inhibition in combination with ACT is an effective strategy to increase efficacy.
- Published
- 2021
22. Adoptive cell therapy targeting common p53 neoantigens in human solid cancers
- Author
-
Steven A. Rosenberg, Zhili Zheng, Yong-Chen Lu, Chuong D. Hoang, Billel Gasmi, Stephanie L. Goff, Paul D. Robbins, Biman C. Paria, Maria R. Parkhurst, Yong Li, Jonathan M. Hernandez, Praveen D. Chatani, Samantha Seitter, Sang-hyun Kim, Drew C. Deniger, Nolan Vale, Maria Florentin, Frank J. Lowery, Sri Krishna, Todd D. Prickett, Sivasish Sindiri, Shoshana Levi, Noam Levin, Victoria Hill, Zhiya Yu, Parisa Malekzadeh, Li Jia, Agnes Y Choi, Elizabeth A. Hedges, Lien Ngo, Jared J. Gartner, Richard M. Sherry, Nikolaos Zacharakis, Satyajit Ray, and James Chih-Hsin Yang
- Subjects
Cell therapy ,business.industry ,Cancer research ,Medicine ,business - Abstract
Adoptive cell therapy (ACT) targeting neoantigens can achieve durable clinical responses in patients with cancer. Most neoantigens arise from rare mutations, requiring highly individualized treatments. To broaden the applicability of ACT targeting neoantigens, we focused on TP53 mutations commonly shared across different cancer types. Here, we describe a library of T cell receptors (TCRs) that can target TP53 mutations shared among 7.3% of patients with solid cancers. These TCRs recognized tumor cells in a TP53 mutation- and human leucocyte antigen (HLA)-specific manner both in vitro and in vivo. Patients with chemorefractory epithelial cancers treated with ex vivo-expanded autologous tumor infiltrating lymphocytes (TILs) naturally reactive with mutant p53 experienced limited clinical responses (2 PRs/12 patients), and we detected low frequencies, exhausted phenotypes, and poor persistence of the infused mutant p53-reactive TILs. Alternatively, we treated one patient with a chemorefractory breast cancer with ACT by transducing autologous peripheral blood lymphocytes with an HLA-A*02-restricted anti-p53R175H TCR. The infused cells exhibited an improved immunophenotype and prolonged persistence compared to the TIL ACT and the patient experienced an objective tumor regression (-55%) that lasted 6 months. Collectively, these data demonstrate the feasibility of off-the-shelf TCR-engineered cell therapies targeting shared p53 neoantigens to treat human cancers.
- Published
- 2021
- Full Text
- View/download PDF
23. Surgical Resection of Pheochromocytomas and Paragangliomas is Associated with Lower Cholesterol Levels
- Author
-
Electron Kebebew, Amy R. Copeland, Meghan L. Good, Naris Nilubol, Parisa Malekzadeh, Shreya Gupta, Dhaval Patel, Karel Pacak, and Samantha M. Ruff
- Subjects
Adult ,Male ,medicine.medical_specialty ,Adolescent ,Urinary system ,Adrenal Gland Neoplasms ,Hyperlipidemias ,Pheochromocytoma ,Gastroenterology ,Article ,Paraganglioma ,Young Adult ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Diabetes mellitus ,Internal medicine ,Hyperlipidemia ,Prevalence ,Humans ,Medicine ,Child ,Prospective cohort study ,Metanephrine ,Aged ,Retrospective Studies ,business.industry ,Cholesterol ,Perioperative ,Middle Aged ,medicine.disease ,chemistry ,030220 oncology & carcinogenesis ,Female ,030211 gastroenterology & hepatology ,Surgery ,business ,Abdominal surgery - Abstract
BACKGROUND: Catecholamine excess in patients with pheochromocytomas or paragangliomas (PPGLs) can lead to hypertension, diabetes and hyperlipidemia. The aim was to investigate the prevalence of hyperlipidemia and the effect of surgical resection. METHODS: One hundred and thirty-two patients with PPGLs underwent an operation at the National Institutes of Health from 2009 to 2016, of which 54 patients met the inclusion criteria. Clinical demographics, BMI, genetic mutations, tumor size, perioperative catecholamine levels and perioperative lipid panels were retrospectively reviewed. Spearman correlation between catecholamines and lipid levels was evaluated. Paired Wilcoxon and paired t test were used to analyze differences in pre- and postoperative lipid levels. RESULTS: Preoperatively, 51 patients (94.4%) had elevated catecholamines, thirteen (24.1%) had elevated total cholesterol (TC) (>200 mg/dL), nine (16.6%) had elevated LDL (>130 mg/dL) and ten (18.5%) had elevated triglycerides (>150 mg/dL). Serum and urinary metanephrine levels were positively associated with TC (r = 0.2792, p = 0.0372 and r = 0.4146, p = 0.0031, respectively) and LDL levels (r = 0.2977, p = 0.0259 and r = 0.4434, p = 0.0014, respectively). Mean TC decreased from 176.4 to 166.3 mg/dL (p = 0.0064) and mean HDL decreased from 56.7 to 53.2 mg/dL (p = 0.0253) after PPGL resection (median 3.1 months (range 1.3–50.2) between lipid panels). Most patients with elevated TC (76.9%) had improvement with mean TC decreasing from 225 to 200.2 mg/dL (p = 0.0230). Of patients with elevated LDL, 66.7% had improvement with mean LDL decreasing from 149 to 131.1 mg/dL (p = 0.0313). CONCLUSIONS: The prevalence of hyperlipidemia in patients with PPGLs is 46%. Future prospective studies are needed to determine whether surgical resection improves TC and/or LDL levels.
- Published
- 2019
- Full Text
- View/download PDF
24. Pilot Trial of Adoptive Transfer of Chimeric Antigen Receptor–transduced T Cells Targeting EGFRvIII in Patients With Glioblastoma
- Author
-
Lori McIntyre, Yong-Chen Lu, Liqiang Xi, Richard A. Morgan, Steven A. Feldman, Stephanie L. Goff, Parisa Malekzadeh, Lily Lu, Howard A. Fine, Richard M. Sherry, Nicholas P. Restifo, Monica Epstein, Zhili Zheng, Paul F. Robbins, Mark Raffeld, James Chih-Hsin Yang, and Steven A. Rosenberg
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Adoptive cell transfer ,Transplantation Conditioning ,T-Lymphocytes ,medicine.medical_treatment ,Immunology ,Receptors, Antigen, T-Cell ,Pilot Projects ,Immunotherapy, Adoptive ,Article ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Interquartile range ,Internal medicine ,Humans ,Immunology and Allergy ,Medicine ,Epidermal growth factor receptor ,Pharmacology ,Chemotherapy ,Receptors, Chimeric Antigen ,biology ,business.industry ,Immunotherapy ,Middle Aged ,Chimeric antigen receptor ,ErbB Receptors ,Treatment Outcome ,030104 developmental biology ,030220 oncology & carcinogenesis ,biology.protein ,Female ,Antibody ,Glioblastoma ,business - Abstract
A deletion variant of epidermal growth factor receptor (EGFRvIII) is a known driver mutation in a subset of primary and secondary glioblastoma multiforme. Adoptive transfer of genetically modified chimeric-antigen receptor (CAR) lymphocytes has demonstrated efficacy in hematologic malignancies but is still early in development for solid cancers. The surface expression of the truncated extracellular ligand domain created by EGFRvIII makes it an attractive target for a CAR-based cancer treatment. Patients with recurrent glioblastoma expressing EGFRvIII were enrolled in a dose escalation phase I trial, using a third-generation chimeric antigen receptor construct derived from a human antibody. Transduced cells were administered after lymphodepleting chemotherapy and supported post-transfer with intravenous interleukin-2. The dose escalation proceeded at half-log increments from 10(7) to >10(10) cells. Primary endpoints were safety and progression-free survival. Eighteen patients were treated with final infusion products ranging from 6.3×10(6) to 2.6×10(10) anti-EGFRvIII-CAR+ T cells. Median progression free survival was 1.3 months (interquartile range 1.1–1.9), with a single outlier of 12.5 months. Two patients experienced severe hypoxia, including one treatment related mortality after cell administration at the highest dose level. All patients developed expected transient hematologic toxicities from preparative chemotherapy. Median overall survival was 6.9 months (interquartile range 2.8–10). Two patients survived over one year, and a third patient was alive at 59 months. Persistence of CAR+ cells correlated with cell dose, but there were no objective responses. Administration of anti-EGFRvIII CAR-transduced T cells did not demonstrate clinically meaningful impact in patients with glioblastoma multiforme in this phase I pilot trial.
- Published
- 2019
- Full Text
- View/download PDF
25. Internal checkpoint regulates T cell neoantigen reactivity and susceptibility to PD1 blockade
- Author
-
Douglas C, Palmer, Beau R, Webber, Yogin, Patel, Matthew J, Johnson, Christine M, Kariya, Walker S, Lahr, Maria R, Parkhurst, Jared J, Gartner, Todd D, Prickett, Frank J, Lowery, Rigel J, Kishton, Devikala, Gurusamy, Zulmarie, Franco, Suman K, Vodnala, Miechaleen D, Diers, Natalie K, Wolf, Nicholas J, Slipek, David H, McKenna, Darin, Sumstad, Lydia, Viney, Tom, Henley, Tilmann, Bürckstümmer, Oliver, Baker, Ying, Hu, Chunhua, Yan, Daoud, Meerzaman, Kartik, Padhan, Winnie, Lo, Parisa, Malekzadeh, Li, Jia, Drew C, Deniger, Shashank J, Patel, Paul F, Robbins, R Scott, McIvor, Modassir, Choudhry, Steven A, Rosenberg, Branden S, Moriarity, and Nicholas P, Restifo
- Subjects
Mice ,Lymphocytes, Tumor-Infiltrating ,T-Lymphocytes ,Animals ,Cytokines ,Humans ,General Medicine ,Adoptive Transfer ,Immunotherapy, Adoptive - Abstract
Adoptive transfer of tumor-infiltrating lymphocytes (TIL) fails to consistently elicit tumor rejection. Manipulation of intrinsic factors that inhibit T cell effector function and neoantigen recognition may therefore improve TIL therapy outcomes. We previously identified the cytokine-induced SH2 protein (CISH) as a key regulator of T cell functional avidity in mice. Here, we investigate the mechanistic role of CISH in regulating human T cell effector function in solid tumors and demonstrate that CRISPR/Cas9 disruption of CISH enhances TIL neoantigen recognition and response to checkpoint blockade.Single-cell gene expression profiling was used to identify a negative correlation between high CISH expression and TIL activation in patient-derived TIL. A GMP-compliant CRISPR/Cas9 gene editing process was developed to assess the impact of CISH disruption on the molecular and functional phenotype of human peripheral blood T cells and TIL. Tumor-specific T cells with disrupted Cish function were adoptively transferred into tumor-bearing mice and evaluated for efficacy with or without checkpoint blockade.CISH expression was associated with T cell dysfunction. CISH deletion using CRISPR/Cas9 resulted in hyper-activation and improved functional avidity against tumor-derived neoantigens without perturbing T cell maturation. Cish knockout resulted in increased susceptibility to checkpoint blockade in vivo.CISH negatively regulates human T cell effector function, and its genetic disruption offers a novel avenue to improve the therapeutic efficacy of adoptive TIL therapy.This study was funded by Intima Bioscience, U.S. and in part through the Intramural program CCR at the National Cancer Institute.
- Published
- 2022
- Full Text
- View/download PDF
26. 152 Adoptive T cell therapy targeting somatic p53 mutations
- Author
-
Elizabeth Hedges, Peter S. Kim, Nolan Vale, Nikolaos Zacharakis, Steven A. Rosenberg, and Parisa Malekzadeh
- Subjects
Adoptive cell transfer ,Colorectal cancer ,business.industry ,Tumor-infiltrating lymphocytes ,T cell ,T-cell receptor ,chemical and pharmacologic phenomena ,Human leukocyte antigen ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,lcsh:RC254-282 ,Metastatic breast cancer ,medicine.anatomical_structure ,Cancer cell ,medicine ,Cancer research ,business - Abstract
Background Adoptive cell therapies (ACT) directed against the products of somatic mutations in cancer cells can lead to long lasting clinical responses. We focused on ACT against shared p53 mutations to be used to potentially treat a broad range of patients with common cancers. We have built a library of anti-mutant p53 T cell receptors (TCRs) to be used for the treatment of patients with epithelial cancers in the autologous setting and as ‘off-the-shelf’ reagents for patients sharing the same p53 mutation and HLA. Methods Tumor infiltrating lymphocytes (TILs) were screened for recognition of p53 mutations and were expanded as previously described.1 For treatment of patient 4349 with metastatic breast cancer, the patient‘s peripheral blood T cells were retrovirally engineered to express the allogeneic anti-p53 R175H TCR. Results We identified TILs recognizing ‘hotspot’ p53 mutations, such as R175H, Y220C, and R273C as well as less frequent but recurrent mutations, such as L111R, C135Y, and Q331H (table 1). First, we adoptively transferred TILs that included T cells reactive to a p53 mutation in an autologous manner for the treatment of patients with metastatic epithelial cancers (n=12). Except for the two patients who exhibited an objective response (RECIST), most of the patients did not respond to the therapy, possibly due to low frequencies of anti-mutant p53 cells in the infusion product, exhausted phenotype, and/or poor persistence (table 2). To overcome these barriers to TIL treatment, we retrovirally transduced autologous peripheral blood T cells to express an allogeneic anti-mutant p53 TCR. We engineered the HLA-A*02:01-restricted anti-p53 R175H TCR into patient 4349’s lymphocytes (transduction efficiency of 64%) and saw less expression of exhaustion markers relative to the TIL infusion products (table 2). This patient with metastatic breast cancer was refractory to the six prior chemotherapy regimens. After the transfer of 5.3e10 cells, the patient experienced an objective partial response, showing regression by 55% of skin and mediastinal lesions for 7 months. The persistence of the infused T cells was higher than the other patients who received the TIL treatment (table 2). Conclusions The library of anti-mutant p53 TCRs we have generated can potentially be used to treat ~6% of all cancer patients. We are pursuing the adoptive transfer of TILs against mutant p53 naturally occurring in the tumor or TCR-engineered cells using ‘off-the-shelf’ receptors against mutant p53. Ethics Approval This study was approved by the Institutional Review Board (IRB) of the NCI, and the approval numbers are as follows:Protocol 10-C-0166 (TIL treatment); Protocol 18-C-0049 (allogeneic TCR engineered T cell therapy) Reference Lo, W., et al., Immunologic Recognition of a Shared p53 Mutated Neoantigen in a Patient with Metastatic Colorectal Cancer. Cancer Immunol Res, 2019. 7(4): p. 534–543.
- Published
- 2020
- Full Text
- View/download PDF
27. Internal checkpoint regulates T cell neoantigen reactivity and susceptibility to PD1 blockade
- Author
-
Modassir Choudhry, Miechaleen D. Diers, Nicholas P. Restifo, Tom Henley, Natalie K. Wolf, Rigel J. Kishton, Paul F. Robbins, Yogin Patel, Lydia Viney, Winnie Lo, Steven A. Rosenberg, Branden S. Moriarity, Tilmann Bürckstümmer, Maria R. Parkhurst, Christine M. Kariya, Parisa Malekzadeh, David H. McKenna, Devikala Gurusamy, Darin Sumstad, Chunhua Yan, Zulmarie Franco, Frank J. Lowery, Douglas C. Palmer, R. Scott McIvor, Daoud Meerzaman, Todd D. Prickett, Matthew Johnson, Ying Hu, Suman K. Vodnala, Nicholas J. Slipek, Shashank J. Patel, Oliver Baker, Li Jia, Drew C. Deniger, Kartik Padhan, Beau R. Webber, Walker S. Lahr, and Jared J. Gartner
- Subjects
Adoptive cell transfer ,Tumor-infiltrating lymphocytes ,T cell ,T-cell receptor ,chemical and pharmacologic phenomena ,Biology ,Cytolysis ,medicine.anatomical_structure ,Cancer research ,biology.protein ,medicine ,Antibody ,CISH ,Protein kinase B - Abstract
While neoantigen-specific tumor infiltrating lymphocytes (TIL) can be derived from in antigen-expressing tumors, their adoptive transfer fails to consistently elicit durable tumor regression. There has been much focus on the role of activation/exhaustion markers such as PD1, CD39 and TOX in TIL senescence. We found these markers were inversely expressed to Cytokine-Induced SH2 protein (CISH), a negative regulator of TCR signaling and tumor immunity in mice. To evaluate the physiological role of CISH in human TIL we developed a high-efficiency CRIPSR-based method to knock out CISH in fully mature TIL. CISH KO resulted in increased T cell receptor (TCR) avidity, tumor cytolysis and neoantigen recognition. CISH expression in the tumor resections correlated with TIL inactivity against p53 hotspot mutations and CISH KO in TIL unmasked reactivity against these universal neoantigens. While CISH KO resulted in T cell hyperactivation and expansion it did not alter maturation, perhaps by preferential PLCγ-1 and not AKT inhibition. Lastly, CISH KO in T cells increased PD1 expression and the adoptive transfer of Cish KO T cells synergistically combines with PD1 antibody blockade resulting in durable tumor regression and survival in a preclinical animal model. These data offer new insights into the regulation of neoantigen recognition, expression of activation/exhaustion markers, and functional/maturation signals in tumor-specific T cells.
- Published
- 2020
- Full Text
- View/download PDF
28. Twenty-Five Year Follow up of a Prospective Randomized Trial Comparing Preoperative vs Postoperative FLAC/Granulocyte-Colony-Stimulating Factor Chemotherapy for Stage II Breast Cancer
- Author
-
Parisa Malekzadeh, Maria J. Merino, Arlene Berman, Kenneth H. Cowan, Craig D. Shriver, Seth M. Steinberg, Meghan L. Good, David N. Danforth, and Kevin Camphausen
- Subjects
Adult ,Cancer Research ,medicine.medical_specialty ,Axillary lymph nodes ,medicine.medical_treatment ,Leucovorin ,Breast Neoplasms ,Article ,Disease-Free Survival ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Randomized controlled trial ,law ,Antineoplastic Combined Chemotherapy Protocols ,Granulocyte Colony-Stimulating Factor ,medicine ,Humans ,030212 general & internal medicine ,Cyclophosphamide ,Aged ,Chemotherapy ,business.industry ,Middle Aged ,medicine.disease ,Primary tumor ,Granulocyte colony-stimulating factor ,Surgery ,Radiation therapy ,medicine.anatomical_structure ,Treatment Outcome ,Oncology ,Fluorouracil ,Chemotherapy, Adjuvant ,Doxorubicin ,030220 oncology & carcinogenesis ,Female ,Radiotherapy, Adjuvant ,business ,medicine.drug ,Follow-Up Studies - Abstract
Background Preoperative chemotherapy is important in the management of women with breast cancer, with the ability to downstage the breast primary tumor and axillary lymph nodes. Long-term studies are needed to identify late toxicities, recurrence patterns, and equivalency with postoperative chemotherapy for recurrence-free survival (RFS) and overall survival (OS). Patients and methods We conducted a single-institution prospective randomized control trial comparing preoperative or postoperative fluorouracil, leucovorin calcium, doxorubicin, and cyclophosphamides/granulocyte colony-stimulating factor chemotherapy for women with untreated clinical stage II (T1N1, T2N0, and T2N1) breast cancer. Long-term follow-up was conducted to define toxicities, recurrence patterns and RFS and OS. Results Fifty-three women with clinical stage II breast cancer were randomized, 26 patients to receive preoperative chemotherapy and 27 to receive postoperative chemotherapy. Long-term follow-up, with a median of 25.3 years, was obtained. Local or systemic recurrence occurred in 8 women in the preoperative group and in 10 women in the postoperative group, and recurrences were predominantly within 10 years of treatment. Late toxicities included local upper extremity paresthesia's, upper extremity edema and congestive heart failure in 1 patient each. Analysis revealed no difference in RFS (20-year RFS probabilities; preoperative: 61.3%, postoperative: 54.7%, P=0.42), or in OS between the 2 treatment groups (20-year probabilities, preoperative: 64.6%, postoperative: 62.2%, P=0.44). Twenty-five of 53 patients (47%) were alive and without disease at this follow-up. Conclusion Twenty-five-year follow-up for this prospective randomized trial confirms the equivalency of preoperative versus postoperative chemotherapy with fluorouracil, leucovorin calcium, doxorubicin, and cyclophosphamides/granulocyte colony-stimulating factor for stage II breast cancer for both RFS and OS.
- Published
- 2020
29. T-cell Responses to TP53 'Hotspot' Mutations and Unique Neoantigens Expressed by Human Ovarian Cancers
- Author
-
Steven A. Rosenberg, Rami Yossef, Anna Pasetto, Jared J. Gartner, David N. Danforth, Li Jia, Parisa Malekzadeh, John R. Wunderlich, Biman C. Paria, Drew C. Deniger, Michelle M. Langhan, Robert Somerville, Paul F. Robbins, and Todd D. Prickett
- Subjects
0301 basic medicine ,Cancer Research ,Somatic cell ,T cell ,Human leukocyte antigen ,Biology ,medicine.disease ,Epitope ,Cell therapy ,Gene expression profiling ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Antigen ,Cancer research ,medicine ,Ovarian cancer - Abstract
Purpose: This was a study prospectively evaluating intratumoral T-cell responses to autologous somatic mutated neoepitopes expressed by human metastatic ovarian cancers. Patients and Methods: Tumor-infiltrating lymphocytes (TIL) were expanded from resected ovarian cancer metastases, which were analyzed by whole-exome and transcriptome sequencing to identify autologous somatic mutations. All mutated neoepitopes, independent of prediction algorithms, were expressed in autologous antigen-presenting cells and then cocultured with TIL fragment cultures. Secretion of IFNγ or upregulation of 41BB indicated a T-cell response. Results: Seven women with metastatic ovarian cancer were evaluated, and 5 patients had clear, dominant T-cell responses to mutated neoantigens, which were corroborated by comparison with the wild-type sequence, identification of the minimal epitope, human leukocyte antigen (HLA) restriction element(s), and neoantigen-specific T-cell receptor(s). Mutated neoantigens were restricted by HLA-B, -C, -DP, -DQ, and/or -DR alleles and appeared to principally arise from random, somatic mutations unique to each patient. We established that TP53 “hotspot” mutations (c.659A>G; p.Y220C and c.733G>A; p.G245S) expressed by two different patients' tumors were both immunogenic in the context of HLA-DRB3*02:02. Conclusions: Mutation-reactive T cells infiltrated ovarian cancer metastases at sufficient frequencies to warrant their investigation as adoptive cell therapy. In addition, transfer of TP53 “hotspot” mutation-reactive T-cell receptors into peripheral blood T cells could be evaluated as a gene therapy for a diverse range of tumor histologies. Clin Cancer Res; 24(22); 5562–73. ©2018 AACR. See related commentary by McNeish, p. 5493
- Published
- 2018
- Full Text
- View/download PDF
30. Outcomes of Adoptive Cell Transfer With Tumor-infiltrating Lymphocytes for Metastatic Melanoma Patients With and Without Brain Metastases
- Author
-
Udai S. Kammula, James Chih-Hsin Yang, Steven A. Rosenberg, John A. Butman, Richard M. Sherry, Gautam U. Mehta, Parisa Malekzadeh, Thomas E. Shelton, Stephanie L. Goff, and Donald E. White
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Adoptive cell transfer ,Skin Neoplasms ,Adolescent ,medicine.medical_treatment ,Immunology ,Immunotherapy, Adoptive ,Article ,Cell therapy ,Young Adult ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,Immunology and Allergy ,Neoplasm Metastasis ,Melanoma ,Aged ,Neoplasm Staging ,Retrospective Studies ,Pharmacology ,Chemotherapy ,Brain Neoplasms ,business.industry ,Tumor-infiltrating lymphocytes ,Immunotherapy ,Middle Aged ,medicine.disease ,Treatment Outcome ,030104 developmental biology ,Tumor progression ,030220 oncology & carcinogenesis ,Cutaneous melanoma ,Female ,business ,Follow-Up Studies - Abstract
Brain metastases cause significant morbidity and mortality in patients with metastatic melanoma. Although adoptive cell therapy (ACT) with tumor-infiltrating lymphocytes (TIL) can achieve complete and durable remission of advanced cutaneous melanoma, the efficacy of this therapy for brain metastases is unclear. Records of patients with M1c melanoma treated with ACT using TIL, including patients with treated and untreated brain metastases, were analyzed. Treatment consisted of preparative chemotherapy, autologous TIL infusion, and high-dose interleukin-2. Treatment outcomes, sites of initial tumor progression, and overall survival were analyzed. Among 144 total patients, 15 patients with treated and 18 patients with untreated brain metastases were identified. Intracranial objective responses (OR) occurred in 28% patients with untreated brain metastases. The systemic OR rates for patients with M1c disease without identified brain disease, treated brain disease, and untreated brain disease, and were 49%, 33% and 33%, respectively, of which 59%, 20% and 16% were durable at last follow-up. The site of untreated brain disease was the most likely site of initial tumor progression (61%) in patients with untreated brain metastases. Overall, we found that ACT with TIL can eliminate small melanoma brain metastases. However, following TIL therapy these patients frequently progress in the brain at a site of untreated brain disease. Patients with treated or untreated brain disease are less likely to achieve durable systemic ORs following TIL therapy compared with M1c disease and no history of brain disease. Melanoma brain metastases likely require local therapy despite the systemic effect of ACT.
- Published
- 2018
- Full Text
- View/download PDF
31. Intrahepatic cholangiocarcinoma as a rare secondary malignancy after allogeneic hematopoietic stem cell transplantation for childhood acute lymphoblastic leukemia: A case report
- Author
-
Nirali N. Shah, Jeremy L. Davis, David E. Kleiner, Meghan L. Good, Parisa Malekzadeh, Jonathan M. Hernandez, Katherine R. Calvo, and Isaac R. Kriley
- Subjects
Transplantation ,medicine.medical_specialty ,business.industry ,Bile duct ,medicine.medical_treatment ,030232 urology & nephrology ,Hematopoietic stem cell transplantation ,030230 surgery ,Malignancy ,medicine.disease ,Gastroenterology ,Article ,Surgical pathology ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Internal medicine ,Pediatrics, Perinatology and Child Health ,Medicine ,Lymphadenectomy ,Hepatectomy ,business ,Childhood Acute Lymphoblastic Leukemia ,Intrahepatic Cholangiocarcinoma - Abstract
Secondary malignancies are a significant cause of non-relapse mortality in patients who undergo allogeneic HCT. However, secondary liver cancer is rare, and ICC following HCT has never been reported in the literature. Secondary solid cancers typically have a long latency period, and cholangiocarcinoma is classically a malignancy occurring in older individuals. Here, we report the first case of secondary ICC, which presented just 3 years after HCT in a young adult with a history of childhood ALL. A 26-year-old male with history of precursor B-cell ALL presented with asymptomatic elevated liver function tests 3 years after HCT. Laboratories were indicative of biliary obstruction. ERCP showed focal biliary stricturing of the common and left hepatic ducts. MRCP revealed left intrahepatic duct dilatation, suggestive of intrahepatic obstructing mass. Additional workup lead to a clinical diagnosis of ICC. The patient underwent left hepatectomy with extrahepatic bile duct resection and portal lymphadenectomy. Surgical pathology was consistent with moderately differentiated cholangiocarcinoma. Our case illustrates a rare SMN following HCT for ALL. It is the first case report of ICC occurring as a secondary cancer in this patient population. Although cholangiocarcinoma is characteristically diagnosed in the older population, it must remain on the differential for biliary obstruction in post-HCT patients.
- Published
- 2020
32. Internal Checkpoint Regulates T Cell Neoantigen Reactivity and Susceptibility to PD1 Blockade
- Author
-
Drew C. Deniger, Rigel J. Kishton, Frank J. Lowery, Ying Hu, Tilmann Bürckstümmer, Yogin Patel, Modassir Choudhry, Matthew D. Johnson, Natalie K. Wolf, Kartik Padhan, Beau R. Webber, Parisa Malekzadeh, David H. McKenna, Paul D. Robbins, Nicholas P. Restifo, Devikala Gurusamy, Steven A. Rosenberg, Tom Henley, Darin Sumstad, Maria R. Parkhurst, Chunhua Yan, Li Jia, Winifred Lo, Zulmarie Franco, Nicholas J. Slipek, R. Scott McIvor, Shashank J. Patel, Oliver Baker, Todd D. Prickett, Lydia Viney, Jared J. Gartner, Suman K. Vodnala, Miechaleen D. Diers, Walker S. Lahr, Douglas C. Palmer, Daoud Meerzaman, Branden S. Moriarity, and Christine M. Kariya
- Subjects
Adoptive cell transfer ,Tumor-infiltrating lymphocytes ,medicine.medical_treatment ,T cell ,T-cell receptor ,chemical and pharmacologic phenomena ,Immunotherapy ,Biology ,Cytolysis ,medicine.anatomical_structure ,medicine ,Cancer research ,CISH ,Protein kinase B - Abstract
While neoantigen-specific tumor infiltrating lymphocytes (TIL) can be derived from in antigen-expressing tumors, their adoptive transfer fails to consistently elicit durable tumor regression. There has been much focus on the role of activation/exhaustion markers such as PD1, CD39 and TOX in TIL senescence. We found these markers were inversely expressed to Cytokine-Induced SH2 protein (CISH), a negative regulator of TCR signaling and tumor immunity in mice. To evaluate the physiological role of CISH in human TIL we developed a high-efficiency CRIPSR-based method to knock out CISH in fully mature TIL. CISH KO resulted in increased T cell receptor (TCR) avidity, tumor cytolysis and neoantigen recognition. CISH expression in the tumor resections correlated with TIL inactivity against p53 hotspot mutations and CISH KO in TIL unmasked reactivity against these universal neoantigens. While CISH KO resulted in T cell hyperactivation and expansion it did not alter maturation, perhaps by preferential PLCγ-1 and not AKT inhibition. Lastly, CISH KO in T cells increased PD1 expression and the adoptive transfer of Cish KO T cells synergistically combines with PD1 antibody blockade resulting in durable tumor regression and survival in a preclinical animal model. These data offer new insights into the regulation of neoantigen recognition, expression of activation/exhaustion markers, and functional/maturation signals in tumor-specific T cells.
- Published
- 2020
- Full Text
- View/download PDF
33. Genome-wide profiling of druggable active tumor defense mechanisms to enhance cancer immunotherapy
- Author
-
Tori N. Yamamoto, Nicholas P. Restifo, Devikala Gurusamy, Zhiya Yu, Amy K Decker, Rigel J. Kishton, Parisa Malekzadeh, Suman K. Vodnala, Drew C. Deniger, Madhusudhanan Sukumar, Douglas C. Palmer, Yogin Patel, Neville E. Sanjana, Michelle Ji, Winifred Lo, Kris C. Wood, Amanda N. Henning, Shashank J. Patel, and Anna Pasetto
- Subjects
0303 health sciences ,Chemokine ,Tumor microenvironment ,biology ,medicine.medical_treatment ,T cell ,Antigen presentation ,Druggability ,Immunotherapy ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Cancer immunotherapy ,030220 oncology & carcinogenesis ,medicine ,biology.protein ,Cancer research ,ITGAV ,030304 developmental biology - Abstract
SummaryAll current highly effective anti-tumor immunotherapeutics depend on the activity of T cells, but tumor cells can escape immune recognition by several mechanisms including loss of function in antigen presentation and inflammatory response genes, expression of immunomodulatory proteins and an immunosuppressive tumor microenvironment. In contrast, the comprehensive identification of strategies that sensitize tumor cells to immunotherapy in vivo has remained challenging. Here, we combine a two-cell type (2CT) whole-genome CRISPR-Cas9 screen with dynamic transcriptional analysis (DTA) of tumor upon T cell encounter to identify a set of genes that tumor cells express as an active defense against T cell-mediated killing. We then employed small molecule and biologic screens designed to antagonize gene products employed by tumor cells to actively defend against T cell-mediated tumor destruction and found that the inhibition of BIRC2, ITGAV or DNPEP enhanced tumor cell destruction by T cells. Mechanistically, we found that BIRC2 promoted immunotherapy resistance through inhibiting non-canonical NF-κB signaling and limiting inflammatory chemokine production. These findings show the path forward to improving T cell-mediated tumor destruction in the clinic.
- Published
- 2019
- Full Text
- View/download PDF
34. Using Human Induced Pluripotent Stem Cells for the Generation of Tumor Antigen-specific T Cells
- Author
-
Parisa Malekzadeh, Meghan L. Good, Hiroshi Kawamoto, Takuya Maeda, Raul Vizcardo, Naritaka Tamaoki, and Nicholas P. Restifo
- Subjects
0301 basic medicine ,Adoptive cell transfer ,General Immunology and Microbiology ,Cellular differentiation ,T cell ,General Chemical Engineering ,General Neuroscience ,T-Lymphocytes ,T-cell receptor ,Induced Pluripotent Stem Cells ,Cell Differentiation ,Biology ,Tumor antigen ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,T cell differentiation ,Cancer research ,medicine ,Humans ,Induced pluripotent stem cell ,Reprogramming ,030215 immunology - Abstract
The generation and expansion of functional T cells in vitro can lead to a broad range of clinical applications. One such use is for the treatment of patients with advanced cancer. Adoptive T cell transfer (ACT) of highly enriched tumor antigen-specific T cells has been shown to cause durable regression of metastatic cancer in some patients. However, during expansion, these cells may become exhausted or senescent, limiting their effector function and persistence in vivo. Induced pluripotent stem cell (iPSC) technology may overcome these obstacles by leading to in vitro generation of large numbers of less differentiated tumor antigen-specific T cells. Human iPSC (hiPSC) have the capacity to differentiate into any type of somatic cell, including lymphocytes, which retain the original T cell receptor (TCR) genomic rearrangement when a T cell is used as a starting cell. Therefore, reprogramming of human tumor antigen-specific T cells to hiPSC followed by redifferentiation to T cell lineage has the potential to produce rejuvenated tumor antigen-specific T cells. Described here is a method for generating tumor antigen-specific CD8αβ+ single positive (SP) T cells from hiPSC using OP9/DLL1 co-culture system. This method is a powerful tool for in vitro T cell lineage generation and will facilitate the development of in vitro derived T cells for use in regenerative medicine and cell-based therapies.
- Published
- 2019
35. Immunologic Recognition of a Shared p53 Mutated Neoantigen in a Patient with Metastatic Colorectal Cancer
- Author
-
Todd D. Prickett, Eric Tran, Anna Pasetto, Scott Kivitz, Steven A. Rosenberg, Paul F. Robbins, Drew C. Deniger, Parisa Malekzadeh, Isaac R. Kriley, Yong-Chen Lu, Maria R. Parkhurst, David S. Schrump, Satyajit Ray, Winifred Lo, Jared J. Gartner, Biman C. Paria, Li Jia, and Thomas E. Shelton
- Subjects
0301 basic medicine ,Adult ,Cancer Research ,Colorectal cancer ,Immunology ,Cell ,Human leukocyte antigen ,Epitope ,Article ,Cell therapy ,03 medical and health sciences ,0302 clinical medicine ,Retrovirus ,Lymphocytes, Tumor-Infiltrating ,Antigen ,Antigens, Neoplasm ,medicine ,Humans ,biology ,HLA-A Antigens ,business.industry ,medicine.disease ,biology.organism_classification ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Female ,Tumor Suppressor Protein p53 ,Ovarian cancer ,business ,Colorectal Neoplasms - Abstract
Adoptive cell therapy (ACT) with T cells targeting neoantigens can mediate durable responses in patients with metastatic cancer. Cell therapies targeting common shared antigens for epithelial cancers are not yet broadly available. Here, we report the identification and characterization in one patient of T-cell receptors (TCRs) recognizing mutated p53 p.R175H, which is shared among a subset of patients with cancer. Tumor-infiltrating lymphocytes were screened for recognition of mutated neoantigens in a patient with metastatic colorectal cancer. HLA-A*0201–restricted recognition of mutated p53 p.R175H was identified, and the minimal peptide epitope was HMTEVVRHC. Reactive T cells were isolated by tetramer sorting, and three TCRs were identified. These TCRs mediated recognition of commercially available ovarian cancer, uterine carcinoma, and myeloma cell lines, as well as an NIH patient–derived esophageal adenocarcinoma line that endogenously expressed p53 p.R175H and HLA-A*0201. They also mediated recognition of p53 p.R175H+ colon, breast, and leukemia cell lines after transduction with a retrovirus encoding HLA-A*0201. This work demonstrates that common shared mutated epitopes such as those found in p53 can elicit immunogenic responses and that the application of ACT may be extended to patients with any cancer histology that expresses both HLA-A*0201 and the p53 p.R175H mutation.
- Published
- 2019
36. Unique Neoantigens Arise from Somatic Mutations in Patients with Gastrointestinal Cancers
- Author
-
John R. Wunderlich, Paul F. Robbins, Anna Pasetto, Li Jia, Mona El-Gamil, Gal Cafri, Maria R. Parkhurst, Eric Tran, Yong F. Li, Lien T. Ngo, Winifred Lo, Robert Somerville, Mojgan Ahmadzadeh, Jessica S. Crystal, Todd D. Prickett, Gabriel Ivey, Satyajit Ray, Frank J. Lowery, Rami Yossef, Abraham Sachs, Steven A. Rosenberg, Almin Lalani, Scott Kivitz, Eric M. Groh, Drew C. Deniger, Parisa Malekzadeh, Stephanie L. Goff, and Jared J. Gartner
- Subjects
0301 basic medicine ,Somatic cell ,medicine.medical_treatment ,T cell ,Receptors, Antigen, T-Cell ,medicine.disease_cause ,Lymphocyte Activation ,Article ,03 medical and health sciences ,0302 clinical medicine ,Lymphocytes, Tumor-Infiltrating ,Antigen ,Antigens, Neoplasm ,T-Lymphocyte Subsets ,medicine ,Biomarkers, Tumor ,Humans ,Gene ,Exome sequencing ,Gastrointestinal Neoplasms ,Mutation ,Tumor-infiltrating lymphocytes ,business.industry ,Immunotherapy ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Disease Susceptibility ,business - Abstract
Immunotherapies can mediate regression of human tumors with high mutation rates, but responses are rarely observed in patients with common epithelial cancers. This raises the question of whether patients with these common cancers harbor T lymphocytes that recognize mutant proteins expressed by autologous tumors that may represent ideal targets for immunotherapy. Using high-throughput immunologic screening of mutant gene products identified via whole-exome sequencing, we identified neoantigen-reactive tumor-infiltrating lymphocytes (TIL) from 62 of 75 (83%) patients with common gastrointestinal cancers. In total, 124 neoantigen-reactive TIL populations were identified, and all but one of the neoantigenic determinants were unique. The results of in vitro T-cell recognition assays demonstrated that 1.6% of the gene products encoded by somatic nonsynonymous mutations were immunogenic. These findings demonstrate that the majority of common epithelial cancers elicit immune recognition and open possibilities for cell-based immunotherapies for patients bearing these cancers. Significance: TILs cultured from 62 of 75 (83%) patients with gastrointestinal cancers recognized neoantigens encoded by 1.6% of somatic mutations expressed by autologous tumor cells, and 99% of the neoantigenic determinants appeared to be unique and not shared between patients. This article is highlighted in the In This Issue feature, p. 983
- Published
- 2018
37. 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
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.