122 results on '"Douglas C Palmer"'
Search Results
2. 620 Multimodal real world data reveals immunogenomic drivers of acquired and primary resistance to immune checkpoint blockade
- Author
-
Ross Stewart, Mark Cobbold, Scott Hammond, Sajan Khosla, Roy Rabbie, Douglas C Palmer, Mohamed Reda Keddar, Sebastian Carrasco Pro, Kathleen Burke, Ana Stewart, Benjamin Sidders, Jonathan R Dry, and Martin L Miller
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2023
- Full Text
- View/download PDF
3. 886 In vitro study of the effects of ceralasertib (AZD6738) on the tumor microenvironment cell populations
- Author
-
Gemma Jones, Luis Vence, Patricia McCoon, Giulia Fabbri, Marta Milo, Douglas C Palmer, Radhika Rayanki, Tao Deng, Mohamed Moustafa, Sonia Iyer, Lydia Greenlees, Steve Arbitman, Arthur Lambert, and Raffaello Cimbro
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2023
- Full Text
- View/download PDF
4. 1169 AZD5863: a specific, potent, affinity-optimized claudin 18.2 and CD3 binding T cell-engager that elicits low cytokine release and is capable of bystander killing
- Author
-
Rakesh Kumar, Yun He, Simon Dovedi, Mark Cobbold, Kristen Pollizzi, Saso Cemerski, Scott Hammond, Marina Natoli, Yiping Rong, Jonathan Fitzgerald, Mary McFarlane, Kathy Mulgrew, Miguel Gaspar, Christopher Lloyd, Douglas C Palmer, Maria AS Broggi, Laure Castan, Sharif Rahmy, Cathryn Kelton, Martin Korade, Oisin Huhn, D Gareth Rees, Anna Sigurdardottir, Benjamin Ridgway, Kathryn Ball, Fred Arce Vargas, Natalia Ceaicovscaia, Gayle Pouliot, and Philip Szekeres
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2023
- Full Text
- View/download PDF
5. Identification of the genomic insertion site of Pmel-1 TCR α and β transgenes by next-generation sequencing.
- Author
-
Yun Ji, Natalie Abrams, Wei Zhu, Eddie Salinas, Zhiya Yu, Douglas C Palmer, Parthav Jailwala, Zulmarie Franco, Rahul Roychoudhuri, Eric Stahlberg, Luca Gattinoni, and Nicholas P Restifo
- Subjects
Medicine ,Science - Abstract
The pmel-1 T cell receptor transgenic mouse has been extensively employed as an ideal model system to study the mechanisms of tumor immunology, CD8+ T cell differentiation, autoimmunity and adoptive immunotherapy. The 'zygosity' of the transgene affects the transgene expression levels and may compromise optimal breeding scheme design. However, the integration sites for the pmel-1 mouse have remained uncharacterized. This is also true for many other commonly used transgenic mice created before the modern era of rapid and inexpensive next-generation sequencing. Here, we show that whole genome sequencing can be used to determine the exact pmel-1 genomic integration site, even with relatively 'shallow' (8X) coverage. The results were used to develop a validated polymerase chain reaction-based genotyping assay. For the first time, we provide a quick and convenient polymerase chain reaction method to determine the dosage of pmel-1 transgene for this freely and publically available mouse resource. We also demonstrate that next-generation sequencing provides a feasible approach for mapping foreign DNA integration sites, even when information of the original vector sequences is only partially known.
- Published
- 2014
- Full Text
- View/download PDF
6. Viral sequestration of antigen subverts cross presentation to CD8(+) T cells.
- Author
-
Eric F Tewalt, Jean M Grant, Erica L Granger, Douglas C Palmer, Neal D Heuss, Dale S Gregerson, Nicholas P Restifo, and Christopher C Norbury
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Virus-specific CD8(+) T cells (T(CD8+)) are initially triggered by peptide-MHC Class I complexes on the surface of professional antigen presenting cells (pAPC). Peptide-MHC complexes are produced by two spatially distinct pathways during virus infection. Endogenous antigens synthesized within virus-infected pAPC are presented via the direct-presentation pathway. Many viruses have developed strategies to subvert direct presentation. When direct presentation is blocked, the cross-presentation pathway, in which antigen is transferred from virus-infected cells to uninfected pAPC, is thought to compensate and allow the generation of effector T(CD8+). Direct presentation of vaccinia virus (VACV) antigens driven by late promoters does not occur, as an abortive infection of pAPC prevents production of these late antigens. This lack of direct presentation results in a greatly diminished or ablated T(CD8+) response to late antigens. We demonstrate that late poxvirus antigens do not enter the cross-presentation pathway, even when identical antigens driven by early promoters access this pathway efficiently. The mechanism mediating this novel means of viral modulation of antigen presentation involves the sequestration of late antigens within virus factories. Early antigens and cellular antigens are cross-presented from virus-infected cells, as are late antigens that are targeted to compartments outside of the virus factories. This virus-mediated blockade specifically targets the cross-presentation pathway, since late antigen that is not cross-presented efficiently enters the MHC Class II presentation pathway. These data are the first to describe an evasion mechanism employed by pathogens to prevent entry into the cross-presentation pathway. In the absence of direct presentation, this evasion mechanism leads to a complete ablation of the T(CD8+) response and a potential replicative advantage for the virus. Such mechanisms of viral modulation of antigen presentation must also be taken into account during the rational design of antiviral vaccines.
- Published
- 2009
- Full Text
- View/download PDF
7. TSCOT+ thymic epithelial cell-mediated sensitive CD4 tolerance by direct presentation.
- Author
-
Sejin Ahn, Gwanghee Lee, Soo Jung Yang, Deokjae Lee, Seunghyuk Lee, Hyo Sun Shin, Min Cheol Kim, Kee Nyung Lee, Douglas C Palmer, Marc R Theoret, Eric J Jenkinson, Graham Anderson, Nicholas P Restifo, and Moon Gyo Kim
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Although much effort has been directed at dissecting the mechanisms of central tolerance, the role of thymic stromal cells remains elusive. In order to further characterize this event, we developed a mouse model restricting LacZ to thymic stromal cotransporter (TSCOT)-expressing thymic stromal cells (TDLacZ). The thymus of this mouse contains approximately 4,300 TSCOT+ cells, each expressing several thousand molecules of the LacZ antigen. TSCOT+ cells express the cortical marker CDR1, CD40, CD80, CD54, and major histocompatibility complex class II (MHCII). When examining endogenous responses directed against LacZ, we observed significant tolerance. This was evidenced in a diverse T cell repertoire as measured by both a CD4 T cell proliferation assay and an antigen-specific antibody isotype analysis. This tolerance process was at least partially independent of Autoimmune Regulatory Element gene expression. When TDLacZ mice were crossed to a novel CD4 T cell receptor (TCR) transgenic reactive against LacZ (BgII), there was a complete deletion of double-positive thymocytes. Fetal thymic reaggregate culture of CD45- and UEA-depleted thymic stromal cells from TDLacZ and sorted TCR-bearing thymocytes excluded the possibility of cross presentation by thymic dendritic cells and medullary epithelial cells for the deletion. Overall, these results demonstrate that the introduction of a neoantigen into TSCOT-expressing cells can efficiently establish complete tolerance and suggest a possible application for the deletion of antigen-specific T cells by antigen introduction into TSCOT+ cells.
- Published
- 2008
- Full Text
- View/download PDF
8. A T cell resilience model associated with response to immunotherapy in multiple tumor types
- Author
-
Yu Zhang, Trang Vu, Douglas C. Palmer, Rigel J. Kishton, Lanqi Gong, Jiao Huang, Thanh Nguyen, Zuojia Chen, Cari Smith, Ferenc Livák, Rohit Paul, Chi-Ping Day, Chuan Wu, Glenn Merlino, Kenneth Aldape, Xin-yuan Guan, and Peng Jiang
- Subjects
Mice ,Receptors, Chimeric Antigen ,Animals ,Humans ,Membrane Proteins ,Immunotherapy ,General Medicine ,CD8-Positive T-Lymphocytes ,Carrier Proteins ,Immunotherapy, Adoptive ,Melanoma ,Article ,General Biochemistry, Genetics and Molecular Biology - Abstract
Despite breakthroughs in cancer immunotherapy, most tumor-reactive T cells cannot persist in solid tumors due to an immunosuppressive environment. We developed Tres (tumor-resilient T cell, https://resilience.ccr.cancer.gov/), a computational model utilizing single-cell transcriptomic data to identify signatures of T cells that are resilient to immunosuppressive signals, such as transforming growth factor-β1, tumor necrosis factor-related apoptosis-inducing ligand and prostaglandin E2. Tres reliably predicts clinical responses to immunotherapy in melanoma, lung cancer, triple-negative breast cancer and B cell malignancies using bulk T cell transcriptomic data from pre-treatment tumors from patients who received immune-checkpoint inhibitors (n = 38), infusion products for chimeric antigen receptor T cell therapies (n = 34) and pre-manufacture samples for chimeric antigen receptor T cell or tumor-infiltrating lymphocyte therapies (n = 84). Further, Tres identified FIBP, whose functions are largely unknown, as the top negative marker of tumor-resilient T cells across many solid tumor types. FIBP knockouts in murine and human donor CD8(+) T cells significantly enhanced T cell-mediated cancer killing in in vitro co-cultures. Further, Fibp knockout in murine T cells potentiated the in vivo efficacy of adoptive cell transfer in the B16 tumor model. Fibp knockout T cells exhibit reduced cholesterol metabolism, which inhibits effector T cell function. These results demonstrate the utility of Tres in identifying biomarkers of T cell effectiveness and potential therapeutic targets for immunotherapies in solid tumors.
- Published
- 2022
- Full Text
- View/download PDF
9. Supplementary Figures 1 - 12 and Tables 1 - 11 from Type I Cytokines Synergize with Oncogene Inhibition to Induce Tumor Growth Arrest
- Author
-
Nicholas P. Restifo, Rahul Roychoudhuri, Marcus W. Bosenberg, Mark Raffeld, Chyi-Chia R. Lee, David F. Stroncek, Francesco Marincola, Ena Wang, Hui Liu, Luca Gattinoni, Zulmarie Franco, James J. Morrow, Madhusudhanan Sukumar, Joseph G. Crompton, Christopher A. Klebanoff, Gautam U. Mehta, Ian S. Goldlust, Ken-ichi Hanada, Alena Gros, Yun Ji, Holger Pflicke, Liqiang Xi, Douglas C. Palmer, Melody Roelke-Parker, Zhiya Yu, David Clever, and Nicolas Acquavella
- Abstract
Fig. S1. Establishment of SB-3123 murine melanoma model. Fig. S2. Schematic of primer design for BRAF c.1799T > A (V600E) mutation analysis in SB-3123p murine melanoma cells. Fig. S3. SB-3123p melanoma is highly sensitive to vemurafenib. Fig. S4. BRAFV600E specific effect of vemurafenib upon SB-3123p established melanomas. Fig. S5. Establishment of SB-3123 murine melanoma cells expressing mouse gp100. Fig. S6. Concurrent vemurafenib therapy is required for sustained tumor growth control. Fig. S7. SB-3123 proliferative capacity is reduced when cultured under combinatorial conditions of vemurafenib and IFN-γ/TNF-α cytokines. Fig. S8. Isobologram analysis from our 8x8 screen for the synergistic (black circle) drug combination of vemurafenib + IFN-γ/TNF-α. Fig. S9. IFN-γ and TNF-α combined with vemurafenib induce growth arrest of SB-3123 melanoma cells. Fig. S10. Melanoma proliferative capacity is reduced when cultured under combinatorial conditions of vemurafenib and IFN-γ/TNF-α cytokines in a BRAFV600E dependent manner. Fig. S11. Human melanoma proliferative capacity is reduced when cultured under combinatorial conditions of vemurafenib and IFN-γ/TNF-α cytokines in a BRAFV600E dependent manner. Fig. S12. Gene set enrichment analysis of vemurafenib + IFN-γ/TNF-α dependent gene expression signature enriched in genes induced by IFN-γ/TNF-α treatment of SB3123. Supplementary Table 1 - All Genes, One Way Anova p FDR < 0.05 - Gene Cluster 1. Supplementary Table 2 - All Genes, One Way Anova p FDR < 0.05 - Gene Cluster 2. Supplementary Table 3 - All Genes, One Way Anova p FDR < 0.05 - Gene Cluster 3. Supplementary Table 4 - All Genes, One Way Anova p FDR < 0.05 - Gene Cluster 4. Supplementary Table 5 - All Genes, One Way Anova p FDR < 0.05 - Gene Cluster 5. Table S6. Gene set enrichment analysis of vemurafenib + IFN-γ/TNF-α dependent gene expression signature normalized to the vemurafenib only dependent gene expression signature in SB3123. Table S7. Gene set enrichment analysis of vemurafenib + IFN-γ/TNF-α dependent gene expression signature normalized to the IFN- γ/TNF-α only dependent gene expression signature in SB3123. Table S8. Gene set enrichment analysis of vemurafenib dependent gene expression signature normalized to control treated SB3123. Supplementary Table 9 - IFNy/TNFB induced genes vs Control Treatment, fold change > 2.0, p-value < 0.01. Supplementary Table 10 - Vemurafenib induced genes vs control treatment, fold change > 2.0, p-value < 0.01. Supplementary Table 11 - Vem + IFNg/TNFa vs IFNg/TNFa differentially expressed genes, FC > 1.5.
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Materials and Methods from Type I Cytokines Synergize with Oncogene Inhibition to Induce Tumor Growth Arrest
- Author
-
Nicholas P. Restifo, Rahul Roychoudhuri, Marcus W. Bosenberg, Mark Raffeld, Chyi-Chia R. Lee, David F. Stroncek, Francesco Marincola, Ena Wang, Hui Liu, Luca Gattinoni, Zulmarie Franco, James J. Morrow, Madhusudhanan Sukumar, Joseph G. Crompton, Christopher A. Klebanoff, Gautam U. Mehta, Ian S. Goldlust, Ken-ichi Hanada, Alena Gros, Yun Ji, Holger Pflicke, Liqiang Xi, Douglas C. Palmer, Melody Roelke-Parker, Zhiya Yu, David Clever, and Nicolas Acquavella
- Abstract
Supplementary Materials and Methods
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Figure 1 from Determinants of Successful CD8+ T-Cell Adoptive Immunotherapy for Large Established Tumors in Mice
- Author
-
Nicholas P. Restifo, Steven A. Rosenberg, Steven E. Finkelstein, Christopher D. Scott, Sid P. Kerkar, Zachary A. Borman, Christian S. Hinrichs, Yun Ji, Pawel Muranski, Douglas C. Palmer, Luca Gattinoni, and Christopher A. Klebanoff
- Abstract
PDF file - 51K
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Figure 2 from Determinants of Successful CD8+ T-Cell Adoptive Immunotherapy for Large Established Tumors in Mice
- Author
-
Nicholas P. Restifo, Steven A. Rosenberg, Steven E. Finkelstein, Christopher D. Scott, Sid P. Kerkar, Zachary A. Borman, Christian S. Hinrichs, Yun Ji, Pawel Muranski, Douglas C. Palmer, Luca Gattinoni, and Christopher A. Klebanoff
- Abstract
PDF file - 57K
- Published
- 2023
- Full Text
- View/download PDF
13. Data from Determinants of Successful CD8+ T-Cell Adoptive Immunotherapy for Large Established Tumors in Mice
- Author
-
Nicholas P. Restifo, Steven A. Rosenberg, Steven E. Finkelstein, Christopher D. Scott, Sid P. Kerkar, Zachary A. Borman, Christian S. Hinrichs, Yun Ji, Pawel Muranski, Douglas C. Palmer, Luca Gattinoni, and Christopher A. Klebanoff
- Abstract
Purpose: Adoptive cell transfer (ACT) of tumor infiltrating or genetically engineered T cells can cause durable responses in patients with metastatic cancer. Multiple clinically modifiable parameters can comprise this therapy, including cell dose and phenotype, in vivo antigen restimulation, and common gamma-chain (γc) cytokine support. However, the relative contributions of each these individual components to the magnitude of the antitumor response have yet to be quantified.Experimental Design: To systematically and quantitatively appraise each of these variables, we employed the Pmel-1 mouse model treating large, established B16 melanoma tumors. In addition to cell dose and magnitude of in vivo antigen restimulation, we also evaluated the relative efficacy of central memory (TCM), effector memory (TEM), and stem cell memory (TSCM) subsets on the strength of tumor regression as well as the dose and type of clinically available γc cytokines, including IL-2, IL-7, IL-15, and IL-21.Results: We found that cell dose, T-cell differentiation status, and viral vaccine titer each were correlated strongly and significantly with the magnitude of tumor regression. Surprisingly, although the total number of IL-2 doses was correlated with tumor regression, no significant benefit to prolonged (≥6 doses) administration was observed. Moreover, the specific type and dose of γc cytokine only moderately correlated with response.Conclusion: Collectively, these findings elucidate some of the key determinants of successful ACT immunotherapy for the treatment of cancer in mice and further show that γc cytokines offer a similar ability to effectively drive antitumor T-cell function in vivo. Clin Cancer Res; 17(16); 5343–52. ©2011 AACR.
- Published
- 2023
- Full Text
- View/download PDF
14. Supplemental Figure S1 from Akt Inhibition Enhances Expansion of Potent Tumor-Specific Lymphocytes with Memory Cell Characteristics
- Author
-
Nicholas P. Restifo, Douglas T. Fearon, Steven A. Rosenberg, Christopher A. Klebanoff, Mark S. Sundrud, Pawel Muranski, Luca Gattinoni, Francesco M. Marincola, Ena Wang, Nicolas Acquavella, Anthony Leonardi, Trevor Upham, Ryan D. Michalek, Sid P. Kerkar, Douglas C. Palmer, Zhiya Yu, Ken-ichi Hanada, Eric Tran, Robert L. Eil, Alena Gros, David Clever, Rahul Roychoudhuri, Madhusudhanan Sukumar, and Joseph G. Crompton
- Abstract
Supplemental Figure S1. Clinical strategy to pharmacologically inhibit Akt during ex vivo expansion of tumor-infiltrating lymphocytes (TIL).
- Published
- 2023
- Full Text
- View/download PDF
15. Supplementary Figure 5 from Tumor-Specific CD8+ T Cells Expressing Interleukin-12 Eradicate Established Cancers in Lymphodepleted Hosts
- Author
-
Nicholas P. Restifo, Giorgio Trinchieri, Steven A. Rosenberg, Luca Gattinoni, Richard A. Morgan, Ling Zhang, Zachary A. Borman, Robert N. Reger, Douglas C. Palmer, Zhiya Yu, Luis Sanchez-Perez, Andrea Boni, Andrew Kaiser, Pawel Muranski, and Sid P. Kerkar
- Abstract
Supplementary Figure 5 from Tumor-Specific CD8+ T Cells Expressing Interleukin-12 Eradicate Established Cancers in Lymphodepleted Hosts
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary Figure 4 from Tumor-Specific CD8+ T Cells Expressing Interleukin-12 Eradicate Established Cancers in Lymphodepleted Hosts
- Author
-
Nicholas P. Restifo, Giorgio Trinchieri, Steven A. Rosenberg, Luca Gattinoni, Richard A. Morgan, Ling Zhang, Zachary A. Borman, Robert N. Reger, Douglas C. Palmer, Zhiya Yu, Luis Sanchez-Perez, Andrea Boni, Andrew Kaiser, Pawel Muranski, and Sid P. Kerkar
- Abstract
Supplementary Figure 4 from Tumor-Specific CD8+ T Cells Expressing Interleukin-12 Eradicate Established Cancers in Lymphodepleted Hosts
- Published
- 2023
- Full Text
- View/download PDF
17. Data from Akt Inhibition Enhances Expansion of Potent Tumor-Specific Lymphocytes with Memory Cell Characteristics
- Author
-
Nicholas P. Restifo, Douglas T. Fearon, Steven A. Rosenberg, Christopher A. Klebanoff, Mark S. Sundrud, Pawel Muranski, Luca Gattinoni, Francesco M. Marincola, Ena Wang, Nicolas Acquavella, Anthony Leonardi, Trevor Upham, Ryan D. Michalek, Sid P. Kerkar, Douglas C. Palmer, Zhiya Yu, Ken-ichi Hanada, Eric Tran, Robert L. Eil, Alena Gros, David Clever, Rahul Roychoudhuri, Madhusudhanan Sukumar, and Joseph G. Crompton
- Abstract
Adoptive cell therapy (ACT) using autologous tumor-infiltrating lymphocytes (TIL) results in complete regression of advanced cancer in some patients, but the efficacy of this potentially curative therapy may be limited by poor persistence of TIL after adoptive transfer. Pharmacologic inhibition of the serine/threonine kinase Akt has recently been shown to promote immunologic memory in virus-specific murine models, but whether this approach enhances features of memory (e.g., long-term persistence) in TIL that are characteristically exhausted and senescent is not established. Here, we show that pharmacologic inhibition of Akt enables expansion of TIL with the transcriptional, metabolic, and functional properties characteristic of memory T cells. Consequently, Akt inhibition results in enhanced persistence of TIL after adoptive transfer into an immunodeficient animal model and augments antitumor immunity of CD8 T cells in a mouse model of cell-based immunotherapy. Pharmacologic inhibition of Akt represents a novel immunometabolomic approach to enhance the persistence of antitumor T cells and improve the efficacy of cell-based immunotherapy for metastatic cancer. Cancer Res; 75(2); 296–305. ©2014 AACR.
- Published
- 2023
- Full Text
- View/download PDF
18. Supplementary Figure 2 from Tumor-Specific CD8+ T Cells Expressing Interleukin-12 Eradicate Established Cancers in Lymphodepleted Hosts
- Author
-
Nicholas P. Restifo, Giorgio Trinchieri, Steven A. Rosenberg, Luca Gattinoni, Richard A. Morgan, Ling Zhang, Zachary A. Borman, Robert N. Reger, Douglas C. Palmer, Zhiya Yu, Luis Sanchez-Perez, Andrea Boni, Andrew Kaiser, Pawel Muranski, and Sid P. Kerkar
- Abstract
Supplementary Figure 2 from Tumor-Specific CD8+ T Cells Expressing Interleukin-12 Eradicate Established Cancers in Lymphodepleted Hosts
- Published
- 2023
- Full Text
- View/download PDF
19. Supplementary Figure 7 from Tumor-Specific CD8+ T Cells Expressing Interleukin-12 Eradicate Established Cancers in Lymphodepleted Hosts
- Author
-
Nicholas P. Restifo, Giorgio Trinchieri, Steven A. Rosenberg, Luca Gattinoni, Richard A. Morgan, Ling Zhang, Zachary A. Borman, Robert N. Reger, Douglas C. Palmer, Zhiya Yu, Luis Sanchez-Perez, Andrea Boni, Andrew Kaiser, Pawel Muranski, and Sid P. Kerkar
- Abstract
Supplementary Figure 7 from Tumor-Specific CD8+ T Cells Expressing Interleukin-12 Eradicate Established Cancers in Lymphodepleted Hosts
- Published
- 2023
- Full Text
- View/download PDF
20. Supplementary Figure 3 from Tumor-Specific CD8+ T Cells Expressing Interleukin-12 Eradicate Established Cancers in Lymphodepleted Hosts
- Author
-
Nicholas P. Restifo, Giorgio Trinchieri, Steven A. Rosenberg, Luca Gattinoni, Richard A. Morgan, Ling Zhang, Zachary A. Borman, Robert N. Reger, Douglas C. Palmer, Zhiya Yu, Luis Sanchez-Perez, Andrea Boni, Andrew Kaiser, Pawel Muranski, and Sid P. Kerkar
- Abstract
Supplementary Figure 3 from Tumor-Specific CD8+ T Cells Expressing Interleukin-12 Eradicate Established Cancers in Lymphodepleted Hosts
- Published
- 2023
- Full Text
- View/download PDF
21. Supplementary Figure 6 from Tumor-Specific CD8+ T Cells Expressing Interleukin-12 Eradicate Established Cancers in Lymphodepleted Hosts
- Author
-
Nicholas P. Restifo, Giorgio Trinchieri, Steven A. Rosenberg, Luca Gattinoni, Richard A. Morgan, Ling Zhang, Zachary A. Borman, Robert N. Reger, Douglas C. Palmer, Zhiya Yu, Luis Sanchez-Perez, Andrea Boni, Andrew Kaiser, Pawel Muranski, and Sid P. Kerkar
- Abstract
Supplementary Figure 6 from Tumor-Specific CD8+ T Cells Expressing Interleukin-12 Eradicate Established Cancers in Lymphodepleted Hosts
- Published
- 2023
- Full Text
- View/download PDF
22. Supplementary Figure 1 from Tumor-Specific CD8+ T Cells Expressing Interleukin-12 Eradicate Established Cancers in Lymphodepleted Hosts
- Author
-
Nicholas P. Restifo, Giorgio Trinchieri, Steven A. Rosenberg, Luca Gattinoni, Richard A. Morgan, Ling Zhang, Zachary A. Borman, Robert N. Reger, Douglas C. Palmer, Zhiya Yu, Luis Sanchez-Perez, Andrea Boni, Andrew Kaiser, Pawel Muranski, and Sid P. Kerkar
- Abstract
Supplementary Figure 1 from Tumor-Specific CD8+ T Cells Expressing Interleukin-12 Eradicate Established Cancers in Lymphodepleted Hosts
- Published
- 2023
- Full Text
- View/download PDF
23. 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
24. 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
25. Publisher Correction: A T cell resilience model associated with response to immunotherapy in multiple tumor types
- Author
-
Yu Zhang, Trang Vu, Douglas C. Palmer, Rigel J. Kishton, Lanqi Gong, Jiao Huang, Thanh Nguyen, Zuojia Chen, Cari Smith, Ferenc Livák, Rohit Paul, Chi-Ping Day, Chuan Wu, Glenn Merlino, Kenneth Aldape, Xin-yuan Guan, and Peng Jiang
- Subjects
General Medicine ,General Biochemistry, Genetics and Molecular Biology - Published
- 2022
- Full Text
- View/download PDF
26. Abstract CT126: A phase 2 trial of first-line AZD0171 + durvalumab and chemotherapy (CT) in patients with metastatic pancreatic ductal adenocarcinoma (PDAC) and CD8+ T cell infiltration
- Author
-
Grainne O'Kane, Teresa Macarulla, Fiyinfolu Balogun, Antoine Hollebecque, Matthew J. Reilley, Sreenivasa R. Chandana, Jim Eyles, Oluwaseun Ojo, Philip Overend, Douglas C. Palmer, Nadia Luheshi, Mayukh Das, Antoine Italiano, and Joan Seoane
- Subjects
Cancer Research ,Oncology - Abstract
Background: Leukemia inhibitory factor (LIF) is an immunosuppressive cytokine linked to tumor growth and metastasis. LIF overexpression correlates with poor prognosis and chemoresistance in multiple tumor types including PDAC. Preclinical data show that LIF promotes an immunosuppressive tumor microenvironment, hindering cytotoxic CD8+ T cell recruitment; low T cell infiltration also correlates with mortality in patients with PDAC. Preclinical studies show LIF-blocking antibodies sensitize tumors to PD-1/PD-L1 inhibition, inhibit epithelial-mesenchymal transition, and prolong survival in combination with CT. In patients with PDAC, higher LIF levels correlate with more aggressive pathology, elevated CA19-9 (a PDAC biomarker), and lower response and survival rates. AZD0171 (formerly MSC-1), a first-in-class, humanized, IgG1 monoclonal antibody, binds specifically and potently to LIF, preventing downstream signaling. In a phase 1 dose escalation study (NCT03490669), AZD0171 monotherapy had manageable safety and led to stable disease (SD) in 34.2% of patients with advanced solid tumors across all dose levels. AZD0171 + CT may improve survival outcomes vs CT alone. Based on preclinical data, AZD0171 may function to stimulate antitumor immune response, and combination with the PD-L1 inhibitor durvalumab could prolong that response and overcome peripheral tolerance in patients with metastatic PDAC. Methods: This is a phase 2, open-label, single-arm, multicenter study of AZD0171 + durvalumab and CT in treatment-naive patients with metastatic PDAC (NCT04999969). Eligible patients must have an ECOG performance status 0 or 1, a Gustave Roussy Immune Score 0 or 1, ≥1 measurable target lesion per RECIST v1.1, and confirmed presence of tumoral CD8+ T cells. Patients with central nervous system metastasis, history of leptomeningeal disease or cord compression, a thromboembolic event ≤3 months prior to study treatment, unresolved grade ≥2 toxicities from prior therapy, or a sensitizing mutation or tumor characteristic for which there is a preferred treatment, are excluded. As PDAC is poorly immune infiltrated, a novel clinical trial assay will be used to select for patients who have existing resident CD8+ T cells and may therefore be more likely to respond. About 115 patients will receive intravenous AZD0171, durvalumab and CT until disease progression or unacceptable toxicity. The primary endpoints are safety and overall survival (OS) rate at 12 months. Secondary endpoints include objective response rate, disease control rate (confirmed response or SD ≥16 weeks), duration of response, median progression-free survival (PFS), PFS rate at 4 months, median OS, pharmacokinetics, pharmacodynamics (changes in serum CA19-9 level and tumor CD8+ T cell infiltration) and immunogenicity. The trial is currently recruiting. Citation Format: Grainne O'Kane, Teresa Macarulla, Fiyinfolu Balogun, Antoine Hollebecque, Matthew J. Reilley, Sreenivasa R. Chandana, Jim Eyles, Oluwaseun Ojo, Philip Overend, Douglas C. Palmer, Nadia Luheshi, Mayukh Das, Antoine Italiano, Joan Seoane. A phase 2 trial of first-line AZD0171 + durvalumab and chemotherapy (CT) in patients with metastatic pancreatic ductal adenocarcinoma (PDAC) and CD8+ T cell infiltration [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr CT126.
- Published
- 2022
- Full Text
- View/download PDF
27. 333 Targeting the apical intracellular checkpoint CISH unleashes T cell neoantigen reactivity and effector program
- Author
-
Matthew D. Johnson, Ying Hu, Winnie Lo, Todd D. Prickett, Douglas C. Palmer, Steven A. Rosenberg, R. Scott McIvor, Daoud Meerzaman, Li Jia, Frank J. Lowery, Parisa Malekzadeh, Walker S. Lahr, Modassir Choudhry, Tilmann Bürckstümmer, Maria R. Parkhurst, Rigel J. Kishton, Nicholas P. Restifo, Tom Henley, David H. McKenna, Devikala Gurusamy, Darin Sumstad, Chunhua Yan, Miechaleen D. Diers, Suman K. Vodnala, Branden S. Moriarity, Zulmarie Franco, Lydia Viney, Christine M. Kariya, Kartik Padhan, Yogin Patel, Natalie K. Wolf, Paul D. Robbins, Beau R. Webber, Jared J. Gartner, Drew C. Deniger, Oliver Baker, Nicholas J. Slipek, and Shashank J. Patel
- Subjects
Adoptive cell transfer ,medicine.medical_treatment ,T cell ,T-cell receptor ,Cell ,Biology ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,Immune checkpoint ,Cytokine ,medicine.anatomical_structure ,Cancer research ,medicine ,CISH ,Intracellular - Abstract
Background Neoantigen-specific T cells isolated from tumors have shown promise clinically but fail to consistently elicit durable tumor regression. Expression of the intracellular checkpoint CISH is elevated in human tumor infiltrating lymphocytes (TIL) and has been shown to inhibit neoantigen reactivity in murine TIL. Methods To explore CISH function in human T cells we developed a CRISPR/Cas9-based strategy to knockout (KO) CISH in human T cells with high-efficiency (>90%) and without detectable off-target editing. Results CISH KO in peripheral blood T cells enhanced proliferation, cytokine polyfunctionality, and cytotoxicity in vitro. To determine if CISH KO similarly enhances TIL function, we developed a clinical-scale, GMP-compliant manufacturing process for CISH disruption in primary human TIL. In process validation runs we achieved CISH KO efficiencies >90% without detectable off-target editing while maintaining high viability and expansion. Compared to WT controls, CISH KO in patient-derived TIL demonstrated increased proliferation, T cell receptor (TCR) avidity, neoantigen recognition, and unmasked reactivity to common p53 mutations. Hyperactivation in CISH KO TIL did not increase differentiation, suggesting that CISH KO may uncouple activation and differentiation pathways. Single cell profiling identifies a pattern of CISH expression inverse to key regulators of activation, and CISH KO in human TIL increases PD1 expression. Adoptive transfer of Cish KO T cells synergistically combines with PD1 inhibition resulting in durable tumor regression in mice, highlighting orthogonal dual cell surface and intracellular checkpoint inhibition as a novel combinatorial approach for T cell immunotherapy. Conclusions These pre-clinical data offer new insight into neoantigen recognition and serve as the basis for a recently initiated human clinical trial at the University of Minnesota (NCT04426669) evaluating inhibition of the novel intracellular immune checkpoint CISH in a CRISPR-engineered, neoantigen-specific T cell therapy for solid tumors. Updates from the clinical trial will be highlighted. Trial Registration NCT04426669
- Published
- 2020
- Full Text
- View/download PDF
28. 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
29. Generation of Tumor Antigen-Specific iPSC-Derived Thymic Emigrants Using a 3D Thymic Culture System
- Author
-
Benjamin L. Kidder, Daisuke Yamada, Takuya Maeda, Haruhiko Koseki, Li Jia, Meghan L. Good, Devikala Gurusamy, Zhiya Yu, Fumito Ito, David F. Stroncek, Marta Bosch-Marce, Naritaka Tamaoki, Zied Abdullaev, Nicholas D. Klemen, Nicholas P. Restifo, Amanda N. Henning, Chengyu Liu, Svetlana Pack, SM Rafiqul Islam, Douglas C. Palmer, Raul Vizcardo, Francis A. Flomerfelt, and Michael J. Kruhlak
- Subjects
0301 basic medicine ,Adoptive cell transfer ,Naive T cell ,medicine.medical_treatment ,Induced Pluripotent Stem Cells ,Cell Culture Techniques ,Thymus Gland ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Imaging, Three-Dimensional ,0302 clinical medicine ,medicine ,Humans ,Induced pluripotent stem cell ,lcsh:QH301-705.5 ,Cell Differentiation ,Translation (biology) ,Immunotherapy ,Phenotype ,Tumor antigen ,030104 developmental biology ,lcsh:Biology (General) ,030220 oncology & carcinogenesis ,Cancer research ,CD8 - Abstract
Summary: Induced pluripotent stem cell (iPSC)-derived T cells may provide future therapies for cancer patients, but those generated by current methods, such as the OP9/DLL1 system, have shown abnormalities that pose major barriers for clinical translation. Our data indicate that these iPSC-derived CD8 single-positive T cells are more like CD4+CD8+ double-positive T cells than mature naive T cells because they display phenotypic markers of developmental arrest and an innate-like phenotype after stimulation. We developed a 3D thymic culture system to avoid these aberrant developmental fates, generating a homogeneous subset of CD8αβ+ antigen-specific T cells, designated iPSC-derived thymic emigrants (iTEs). iTEs exhibit phenotypic and functional similarities to naive T cells both in vitro and in vivo, including the capacity for expansion, memory formation, and tumor suppression. These data illustrate the limitations of current methods and provide a tool to develop the next generation of iPSC-based antigen-specific immunotherapies. : A barrier for clinical application of iPSC-derived CD8 T cells using OP9/DLL1 is their abnormal biology. Vizcardo et al. show that a 3D thymic culture system enables the generation of a homogeneous antigen-specific T cell subset, named iTEs, which closely mimics naive T cells and exhibits potent anti-tumor activity. Keywords: thymopoiesis, T cell differentiation, iPSC differentiation, adoptive cell transfer, naïve T cell, recent rhymic emigrants, fetal thymus organ culture, immunotherapy, 3D culture, tumor antigen specific T cell
- Published
- 2018
- Full Text
- View/download PDF
30. 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
31. 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
32. Multi-phenotype CRISPR-Cas9 Screen Identifies p38 Kinase as a Target for Adoptive Immunotherapies
- Author
-
Christine M. Kariya, Li Jia, Tori N. Yamamoto, Shashank J. Patel, Devikala Gurusamy, Sri Krishna, Zhiya Yu, Arash Eidizadeh, Madhusudhanan Sukumar, Amanda N. Henning, Nicholas P. Restifo, Mary A. Black, Nikolaos Zacharakis, Jenny H. Pan, Suman K. Vodnala, Douglas C. Palmer, Robert L. Eil, and Rigel J. Kishton
- Subjects
0301 basic medicine ,Male ,Cancer Research ,DNA damage ,p38 mitogen-activated protein kinases ,T-Lymphocytes ,Regulator ,Melanoma, Experimental ,Receptors, Antigen, T-Cell ,Breast Neoplasms ,Biology ,Immunotherapy, Adoptive ,p38 Mitogen-Activated Protein Kinases ,03 medical and health sciences ,Mice ,0302 clinical medicine ,CRISPR ,Animals ,Mice, Knockout ,Kinase ,Cell Differentiation ,Phenotype ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Female ,NY-ESO-1 ,CRISPR-Cas Systems ,Genetic Engineering ,Genetic screen - Abstract
Summary T cells are central to all currently effective cancer immunotherapies, but the characteristics defining therapeutically effective anti-tumor T cells have not been comprehensively elucidated. Here, we delineate four phenotypic qualities of effective anti-tumor T cells: cell expansion, differentiation, oxidative stress, and genomic stress. Using a CRISPR-Cas9-based genetic screen of primary T cells we measured the multi-phenotypic impact of disrupting 25 T cell receptor-driven kinases. We identified p38 kinase as a central regulator of all four phenotypes and uncovered transcriptional and antioxidant pathways regulated by p38 in T cells. Pharmacological inhibition of p38 improved the efficacy of mouse anti-tumor T cells and enhanced the functionalities of human tumor-reactive and gene-engineered T cells, paving the way for clinically relevant interventions.
- Published
- 2019
33. BACH2 regulates CD8(+) T cell differentiation by controlling access of AP-1 factors to enhancers
- Author
-
Klaus Okkenhaug, Yuka Kanno, Luca Gattinoni, Han-Yu Shih, Peng Li, Giulia Fabozzi, Kylie M. Quinn, Yun Ji, Derek C. Macallan, Jun Zhu, Yoshiyuki Wakabayashi, Shashank J. Patel, Warren J. Leonard, Robert L. Eil, Akihiko Muto, Zhiya Yu, Christopher A. Klebanoff, Kazuhiko Igarashi, Douglas C. Palmer, Rosanne Spolski, Rahul Roychoudhuri, Jenny H. Pan, David Clever, Madhusudhanan Sukumar, Nicholas P. Restifo, John J. O'Shea, Roychoudhuri, Rahul [0000-0002-5392-1853], Okkenhaug, Klaus [0000-0002-9432-4051], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Cellular differentiation ,Immunology ,Oncogene Protein p65(gag-jun) ,Vaccinia virus ,Biology ,Adaptive Immunity ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,Article ,03 medical and health sciences ,Mice ,Vaccinia ,Immunology and Allergy ,Cytotoxic T cell ,Animals ,Enhancer ,Cells, Cultured ,Mice, Knockout ,Effector ,Activator (genetics) ,T-cell receptor ,Cell Differentiation ,Acquired immune system ,Mice, Inbred C57BL ,Transcription Factor AP-1 ,030104 developmental biology ,Basic-Leucine Zipper Transcription Factors ,Enhancer Elements, Genetic ,Gene Expression Regulation ,Cancer research ,Signal transduction ,Immunologic Memory ,Signal Transduction - Abstract
T cell antigen receptor (TCR) signaling drives distinct responses depending upon the differentiation state and context of CD8+ T cells. We hypothesized that access of signal-dependent transcription factors (TFs) to enhancers is dynamically regulated to shape transcriptional responses to TCR signaling. We found that the TF BACH2 restrains terminal differentiation to enable generation of long-lived memory cells and protective immunity following viral infection. BACH2 was recruited to enhancers where it limited expression of TCR-driven genes by attenuating the availability of activator protein 1 (AP-1) sites to Jun family signal-dependent TFs. In naïve cells, this prevented TCR-driven induction of genes associated with terminal differentiation. Upon effector differentiation, reduced expression of BACH2 and its phosphorylation enabled unrestrained induction of TCR-driven effector programs.
- Published
- 2018
- Full Text
- View/download PDF
34. T cell stemness and dysfunction in tumors are triggered by a common mechanism
- Author
-
Min Hwa Shin, Tori N. Yamamoto, Robert L. Eil, Xiaojing Liu, Nicholas P. Restifo, Douglas C. Palmer, Jing Huang, Suman K. Vodnala, Jason W. Locasale, Rigel J. Kishton, Michael J. Kruhlak, Christopher A. Klebanoff, Ngoc Han Ha, Shashank J. Patel, Toren Finkel, Madhusudhanan Sukumar, Ping-Hsien Lee, Rahul Roychoudhuri, and Zhiya Yu
- Subjects
T cell ,medicine.medical_treatment ,Biology ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,Immune tolerance ,Epigenesis, Genetic ,Histones ,Mice ,Lymphocytes, Tumor-Infiltrating ,Cancer immunotherapy ,Acetyl Coenzyme A ,Neoplasms ,medicine ,Autophagy ,Immune Tolerance ,Tumor Microenvironment ,Animals ,Humans ,Caloric Restriction ,Tumor microenvironment ,Multidisciplinary ,Effector ,Stem Cells ,Acetylation ,Cell Differentiation ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Histone ,Cancer research ,biology.protein ,Potassium ,CD8 - Abstract
Stemness against adversity T lymphocytes are powerful immune cells that can destroy tumors, but cancers have developed tricks to evade killing. Vodnala et al. found that potassium ions in the tumor microenvironment serve a dual role of influencing T cell effector function and stemness (see the Perspective by Baixauli Celda et al. ). Increased potassium impairs T cell metabolism and nutrient uptake, resulting in a starvation state known as autophagy. The increased potassium can also preserve T cells in a stem-like state where they retain the capacity to divide. These seemingly divergent processes are linked to the cellular distribution of acetyl–coenzyme A, which, when manipulated, can restore the ability of human T cells to eliminate tumors in mice. Science , this issue p. eaau0135 ; see also p. 1395
- Published
- 2018
35. Mitochondrial Membrane Potential Identifies Cells with Enhanced Stemness for Cellular Therapy
- Author
-
David S. Schrump, Shashank J. Patel, Toren Finkel, Mahadev Rao, Luca Gattinoni, Francesco M. Marincola, Yun Ji, Peng Li, Robert L. Eil, Madhusudhanan Sukumar, David Clever, Jie Liu, Rahul Roychoudhuri, Gautam U. Mehta, Suman Mitra, Zhiya Yu, Pawel Muranski, Nicholas P. Restifo, Ena Wang, Joseph G. Crompton, Greg Whitehill, Douglas C. Palmer, Keyvan Keyvanfar, Christopher A. Klebanoff, and Warren J. Leonard
- Subjects
0301 basic medicine ,Physiology ,T cell ,Melanoma, Experimental ,CD8-Positive T-Lymphocytes ,Biology ,Article ,Cell therapy ,Transcriptome ,03 medical and health sciences ,T-Lymphocyte Subsets ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Molecular Biology ,Membrane Potential, Mitochondrial ,Mice, Knockout ,Cell Biology ,Lymphoid Progenitor Cells ,Hematopoietic Stem Cells ,Cell biology ,Mice, Inbred C57BL ,Oxidative Stress ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,Cell culture ,Cytokines ,Stem cell ,Neoplasm Transplantation ,CD8 ,Stem Cell Transplantation - Abstract
Long-term survival and antitumor immunity of adoptively transferred CD8(+) T cells is dependent on their metabolic fitness, but approaches to isolate therapeutic T cells based on metabolic features are not well established. Here we utilized a lipophilic cationic dye tetramethylrhodamine methyl ester (TMRM) to identify and isolate metabolically robust T cells based on their mitochondrial membrane potential (ΔΨm). Comprehensive metabolomic and gene expression profiling demonstrated global features of improved metabolic fitness in low-ΔΨm-sorted CD8(+) T cells. Transfer of these low-ΔΨm T cells was associated with superior long-term in vivo persistence and an enhanced capacity to eradicate established tumors compared with high-ΔΨm cells. Use of ΔΨm-based sorting to enrich for cells with superior metabolic features was observed in CD8(+), CD4(+) T cell subsets, and long-term hematopoietic stem cells. This metabolism-based approach to cell selection may be broadly applicable to therapies involving the transfer of HSC or lymphocytes for the treatment of viral-associated illnesses and cancer.
- Published
- 2016
- Full Text
- View/download PDF
36. Lineage relationship of CD8+ T cell subsets is revealed by progressive changes in the epigenetic landscape
- Author
-
Douglas C. Palmer, John S. Tsang, Suresh Cuddapah, Christopher A. Klebanoff, Ena Wang, Joseph G. Crompton, Yun Ji, Manikandan Narayanan, Luca Gattinoni, Rahul Roychoudhuri, Keji Zhao, Francesco M. Marincola, Shashank J. Patel, Madhusudhanan Sukumar, Weiqun Peng, Wenjing Yang, and Nicholas P. Restifo
- Subjects
Histone H3 Lysine 4 ,Epigenetics in learning and memory ,Immunology ,CD8-Positive T-Lymphocytes ,Biology ,Methylation ,Chromatin remodeling ,Epigenesis, Genetic ,Histones ,03 medical and health sciences ,0302 clinical medicine ,Animals ,Immunology and Allergy ,Cytotoxic T cell ,Cell Lineage ,Epigenetics ,Promoter Regions, Genetic ,Research Articles ,030304 developmental biology ,Epigenomics ,Genetics ,0303 health sciences ,Gene Expression Profiling ,Chromatin Assembly and Disassembly ,Lymphocyte Subsets ,Mice, Inbred C57BL ,Enhancer Elements, Genetic ,Infectious Diseases ,Histone ,biology.protein ,H3K4me3 ,Immunologic Memory ,Protein Processing, Post-Translational ,030215 immunology - Abstract
To better elucidate epigenetic mechanisms that correlate with the dynamic gene expression program observed upon T-cell differentiation, we investigated the genomic landscape of histone modifications in naive and memory CD8(+) T cells. Using a ChIP-Seq approach coupled with global gene expression profiling, we generated genome-wide histone H3 lysine 4 (H3K4me3) and H3 lysine 27 (H3K27me3) trimethylation maps in naive, T memory stem cells, central memory cells, and effector memory cells in order to gain insight into how histone architecture is remodeled during T cell differentiation. We show that H3K4me3 histone modifications are associated with activation of genes, while H3K27me3 is negatively correlated with gene expression at canonical loci and enhancers associated with T-cell metabolism, effector function, and memory. Our results also reveal histone modifications and gene expression signatures that distinguish the recently identified T memory stem cells from other CD8(+) T-cell subsets. Taken together, our results suggest that CD8(+) lymphocytes undergo chromatin remodeling in a progressive fashion. These findings have major implications for our understanding of peripheral T-cell ontogeny and the formation of immunological memory.
- Published
- 2015
- Full Text
- View/download PDF
37. The Cish SH2 domain is essential for PLC-gamma 1 regulation in TCR stimulated CD8(+) T cells
- Author
-
Nicholas P. Restifo, Ana Dios-Esponera, Geoffrey Guittard, Itoro Akpan, Asit Kumar Manna, Douglas C. Palmer, Valarie A. Barr, Lawrence E. Samelson, Centre de Recherche en Cancérologie de Marseille (CRCM), Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut Paoli-Calmettes, Fédération nationale des Centres de lutte contre le Cancer (FNCLCC)-Fédération nationale des Centres de lutte contre le Cancer (FNCLCC)-Aix Marseille Université (AMU), National Institutes of Health [Bethesda] (NIH), Intramural Research Program of the National Cancer Institute Center for Cancer Research, Aix Marseille Université (AMU)-Institut Paoli-Calmettes, Fédération nationale des Centres de lutte contre le Cancer (FNCLCC)-Fédération nationale des Centres de lutte contre le Cancer (FNCLCC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), and Bidaut, Ghislain
- Subjects
0301 basic medicine ,T cell ,[SDV]Life Sciences [q-bio] ,Receptors, Antigen, T-Cell ,Gene Expression ,lcsh:Medicine ,Suppressor of Cytokine Signaling Proteins ,Cytotoxic T cells ,CD8-Positive T-Lymphocytes ,Signal transduction ,Lymphocyte Activation ,SH2 domain ,Article ,Cell Line ,src Homology Domains ,Mice ,03 medical and health sciences ,medicine ,Animals ,Humans ,Cytotoxic T cell ,Protein Interaction Domains and Motifs ,lcsh:Science ,CISH ,Mice, Knockout ,Multidisciplinary ,biology ,Phospholipase C gamma ,Chemistry ,lcsh:R ,T-cell receptor ,3. Good health ,Ubiquitin ligase ,Cell biology ,[SDV] Life Sciences [q-bio] ,030104 developmental biology ,medicine.anatomical_structure ,biology.protein ,Cytokines ,Calcium ,lcsh:Q ,CD8 ,Protein Binding - Abstract
Cish, participates within a multi-molecular E3 ubiquitin ligase complex, which ubiquitinates target proteins. It has an inhibitory effect on T cell activation mediated by PLC-γ1 regulation, and it functions as a potent checkpoint in CD8+ T cell tumor immunotherapy. To study the structural and functional relationships between Cish and PLC-γ1 during CD8+ T cell activation, we tested mutants of the Cish-SH2 (R107K) and D/BC (L222Q, C226Q) domains. We confirmed that Cish-SH2-specific binding was essential for PLC-γ1 ubiquitination and degradation. This domain was essential for the Cish-mediated inhibition of Ca2+ release upon TCR stimulation. No effect on inhibition of cytokine release was observed with SH2 or D/BC mutants, although the absence of Cish led to an increased release of IFN-γ and TNF-α. Using imaging we showed that Cish was expressed mostly in the cytoplasm and we did not see any Cish clustering at the plasma membrane upon stimulation. We conclude that the Cish-SH2 domain is essential for PLC-γ1 regulation in TCR-stimulated CD8+ T cells.
- Published
- 2018
- Full Text
- View/download PDF
38. Akt Inhibition Enhances Expansion of Potent Tumor-Specific Lymphocytes with Memory Cell Characteristics
- Author
-
Nicolas Acquavella, Robert L. Eil, Anthony J. Leonardi, Alena Gros, Sid P. Kerkar, David Clever, Joseph G. Crompton, Steven A. Rosenberg, Christopher A. Klebanoff, Trevor Upham, Ena Wang, Nicholas P. Restifo, Douglas C. Palmer, Douglas T. Fearon, Ryan D. Michalek, Mark S. Sundrud, Madhusudhanan Sukumar, Luca Gattinoni, Ken-ichi Hanada, Eric Tran, Rahul Roychoudhuri, Zhiya Yu, Pawel Muranski, and Francesco M. Marincola
- Subjects
Cancer Research ,Adoptive cell transfer ,medicine.medical_treatment ,Cell ,Melanoma, Experimental ,Mice, Transgenic ,chemical and pharmacologic phenomena ,Biology ,Immunotherapy, Adoptive ,Article ,Cell therapy ,Mice ,Random Allocation ,Lymphocytes, Tumor-Infiltrating ,Mice, Inbred NOD ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Cytotoxic T cell ,Melanoma ,Protein Kinase Inhibitors ,Protein kinase B ,Cancer ,Immunotherapy ,medicine.disease ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Oncology ,Immunology ,Cancer research ,Immunologic Memory ,Proto-Oncogene Proteins c-akt - Abstract
Adoptive cell therapy (ACT) using autologous tumor-infiltrating lymphocytes (TIL) results in complete regression of advanced cancer in some patients, but the efficacy of this potentially curative therapy may be limited by poor persistence of TIL after adoptive transfer. Pharmacologic inhibition of the serine/threonine kinase Akt has recently been shown to promote immunologic memory in virus-specific murine models, but whether this approach enhances features of memory (e.g., long-term persistence) in TIL that are characteristically exhausted and senescent is not established. Here, we show that pharmacologic inhibition of Akt enables expansion of TIL with the transcriptional, metabolic, and functional properties characteristic of memory T cells. Consequently, Akt inhibition results in enhanced persistence of TIL after adoptive transfer into an immunodeficient animal model and augments antitumor immunity of CD8 T cells in a mouse model of cell-based immunotherapy. Pharmacologic inhibition of Akt represents a novel immunometabolomic approach to enhance the persistence of antitumor T cells and improve the efficacy of cell-based immunotherapy for metastatic cancer. Cancer Res; 75(2); 296–305. ©2014 AACR.
- Published
- 2015
- Full Text
- View/download PDF
39. Cish actively silences TCR signaling in CD8+ T cells to maintain tumor tolerance
- Author
-
Lakshmi Balagopalan, Christopher A. Klebanoff, Geoffrey Guittard, Madhusudhanan Sukumar, Robert L. Eil, Douglas C. Palmer, Zulmarie Franco, Luca Gattinoni, Nicholas P. Restifo, David Clever, Rajat Varma, Ena Wang, Lawrence E. Samelson, Joseph G. Crompton, Yun Ji, Shashank J. Patel, Anna Chichura, Rahul Roychoudhuri, Francesco M. Marincola, and Nicholas J. Van Panhuys
- Subjects
inorganic chemicals ,Mice, 129 Strain ,medicine.medical_treatment ,Immunology ,Immunoblotting ,Melanoma, Experimental ,Receptors, Antigen, T-Cell ,chemical and pharmacologic phenomena ,Suppressor of Cytokine Signaling Proteins ,Biology ,CD8-Positive T-Lymphocytes ,Immunotherapy, Adoptive ,Article ,Immune tolerance ,Cancer immunotherapy ,Cell Line, Tumor ,medicine ,Immune Tolerance ,Tumor Microenvironment ,otorhinolaryngologic diseases ,Immunology and Allergy ,Cytotoxic T cell ,Animals ,Humans ,Avidity ,CISH ,Cells, Cultured ,Research Articles ,Cell Proliferation ,Oligonucleotide Array Sequence Analysis ,Mice, Knockout ,Tumor microenvironment ,Microscopy, Confocal ,Phospholipase C gamma ,Reverse Transcriptase Polymerase Chain Reaction ,T-cell receptor ,Mice, Inbred C57BL ,Cancer research ,sense organs ,Transcriptome ,CD8 ,psychological phenomena and processes ,Protein Binding ,Signal Transduction - Abstract
Palmer et al. find that Cish, a member of the SOCS family, is induced by TCR stimulation in CD8+ T cells and inhibits their functional avidity against tumor. The authors uncover a novel mechanism of suppression for a SOCS member., Improving the functional avidity of effector T cells is critical in overcoming inhibitory factors within the tumor microenvironment and eliciting tumor regression. We have found that Cish, a member of the suppressor of cytokine signaling (SOCS) family, is induced by TCR stimulation in CD8+ T cells and inhibits their functional avidity against tumors. Genetic deletion of Cish in CD8+ T cells enhances their expansion, functional avidity, and cytokine polyfunctionality, resulting in pronounced and durable regression of established tumors. Although Cish is commonly thought to block STAT5 activation, we found that the primary molecular basis of Cish suppression is through inhibition of TCR signaling. Cish physically interacts with the TCR intermediate PLC-γ1, targeting it for proteasomal degradation after TCR stimulation. These findings establish a novel targetable interaction that regulates the functional avidity of tumor-specific CD8+ T cells and can be manipulated to improve adoptive cancer immunotherapy.
- Published
- 2015
40. miR-155 augments CD8 + T-cell antitumor activity in lymphoreplete hosts by enhancing responsiveness to homeostatic γ c cytokines
- Author
-
Nicholas P. Restifo, David Clever, Zulmarie Franco, Yun Ji, Douglas C. Palmer, Nga V. Hawk, Madhusudhanan Sukumar, William G. Telford, Claudia Wrzesinski, Zhiya Yu, Rahul Roychoudhuri, Luca Gattinoni, Sanjivan Gautam, Christopher A. Klebanoff, Charles D. Surh, Thomas A. Waldmann, and Jinhui Hu
- Subjects
medicine.medical_treatment ,Melanoma, Experimental ,Mice, Transgenic ,CD8-Positive T-Lymphocytes ,Immunotherapy, Adoptive ,Mice ,Cell Line, Tumor ,STAT5 Transcription Factor ,medicine ,Animals ,Humans ,Cytotoxic T cell ,Protein kinase B ,STAT5 ,Mice, Knockout ,Multidisciplinary ,Base Sequence ,biology ,Suppressor of cytokine signaling 1 ,Biological Sciences ,Mice, Inbred C57BL ,MicroRNAs ,HEK293 Cells ,Cytokine ,biology.protein ,STAT protein ,Cancer research ,Cytokines ,Signal transduction ,Proto-Oncogene Proteins c-akt ,CD8 ,Signal Transduction ,gp100 Melanoma Antigen - Abstract
Lymphodepleting regimens are used before adoptive immunotherapy to augment the antitumor efficacy of transferred T cells by removing endogenous homeostatic "cytokine sinks." These conditioning modalities, however, are often associated with severe toxicities. We found that microRNA-155 (miR-155) enabled tumor-specific CD8(+) T cells to mediate profound antitumor responses in lymphoreplete hosts that were not potentiated by immune-ablation. miR-155 enhanced T-cell responsiveness to limited amounts of homeostatic γc cytokines, resulting in delayed cellular contraction and sustained cytokine production. miR-155 restrained the expression of the inositol 5-phosphatase Ship1, an inhibitor of the serine-threonine protein kinase Akt, and multiple negative regulators of signal transducer and activator of transcription 5 (Stat5), including suppressor of cytokine signaling 1 (Socs1) and the protein tyrosine phosphatase Ptpn2. Expression of constitutively active Stat5a recapitulated the survival advantages conferred by miR-155, whereas constitutive Akt activation promoted sustained effector functions. Our results indicate that overexpression of miR-155 in tumor-specific T cells can be used to increase the effectiveness of adoptive immunotherapies in a cell-intrinsic manner without the need for life-threatening, lymphodepleting maneuvers.
- Published
- 2014
- Full Text
- View/download PDF
41. Inhibiting glycolytic metabolism enhances CD8+ T cell memory and antitumor function
- Author
-
Christopher A. Klebanoff, Jie Liu, Robert P. Mohney, Madhusudhanan Sukumar, Joseph G. Crompton, Nicholas P. Restifo, Zhiya Yu, Pawel Muranski, Douglas C. Palmer, Ashish Lal, Toren Finkel, Luca Gattinoni, Edward D. Karoly, Yun Ji, Murugan Subramanian, and Rahul Roychoudhuri
- Subjects
Adoptive cell transfer ,T cell ,Glucose uptake ,Melanoma, Experimental ,Glucose analog ,General Medicine ,CD8-Positive T-Lymphocytes ,Biology ,medicine.anatomical_structure ,Biochemistry ,Antigen ,Cell culture ,medicine ,Animals ,Humans ,Cytotoxic T cell ,Glycolysis ,CD8 ,Research Article - Abstract
Naive CD8+ T cells rely upon oxidation of fatty acids as a primary source of energy. After antigen encounter, T cells shift to a glycolytic metabolism to sustain effector function. It is unclear, however, whether changes in glucose metabolism ultimately influence the ability of activated T cells to become long-lived memory cells. We used a fluorescent glucose analog, 2-NBDG, to quantify glucose uptake in activated CD8+ T cells. We found that cells exhibiting limited glucose incorporation had a molecular profile characteristic of memory precursor cells and an increased capacity to enter the memory pool compared with cells taking up high amounts of glucose. Accordingly, enforcing glycolytic metabolism by overexpressing the glycolytic enzyme phosphoglycerate mutase-1 severely impaired the ability of CD8+ T cells to form long-term memory. Conversely, activation of CD8+ T cells in the presence of an inhibitor of glycolysis, 2-deoxyglucose, enhanced the generation of memory cells and antitumor functionality. Our data indicate that augmenting glycolytic flux drives CD8+ T cells toward a terminally differentiated state, while its inhibition preserves the formation of long-lived memory CD8+ T cells. These results have important implications for improving the efficacy of T cell–based therapies against chronic infectious diseases and cancer.
- Published
- 2013
- Full Text
- View/download PDF
42. Collapse of the Tumor Stroma is Triggered by IL-12 Induction of Fas
- Author
-
Jenny H. Pan, Ling Zhang, Nicholas J. Van Panhuys, Nicholas P. Restifo, Richard A. Morgan, Steven A. Rosenberg, Zhiya Yu, Anthony J. Leonardi, Sid P. Kerkar, Douglas C. Palmer, and Joseph G. Crompton
- Subjects
Stromal cell ,Antigen presentation ,Melanoma, Experimental ,Mice, Transgenic ,CD8-Positive T-Lymphocytes ,Biology ,Fas ligand ,Mice ,Interleukin 21 ,Drug Discovery ,Genetics ,Animals ,Cytotoxic T cell ,fas Receptor ,Molecular Biology ,Interleukin 3 ,Pharmacology ,Dendritic Cells ,Flow Cytometry ,Interleukin-12 ,Mice, Inbred C57BL ,Immunology ,Cancer research ,Interleukin 12 ,Molecular Medicine ,Original Article ,Female ,CD8 - Abstract
Engineering CD8⁺ T cells to deliver interleukin 12 (IL-12) to the tumor site can lead to striking improvements in the ability of adoptively transferred T cells to induce the regression of established murine cancers. We have recently shown that IL-12 triggers an acute inflammatory environment that reverses dysfunctional antigen presentation by myeloid-derived cells within tumors and leads to an increase in the infiltration of adoptively transferred antigen-specific CD8⁺ T cells. Here, we find that local delivery of IL-12 increased the expression of Fas within tumor-infiltrating macrophages, dendritic cells, and myeloid-derived suppressor cells (MDSC), and that these changes were abrogated in mice deficient in IL-12-receptor signaling. Importantly, upregulation of Fas in host mice played a critical role in the proliferation and antitumor activity of adoptively transferred IL-12-modified CD8⁺ T cells. We also observed higher percentages of myeloid-derived cell populations within tumors in Fas-deficient mice, indicating that tumor stromal destruction was dependent on the Fas death receptor. Taken together, these results describe the likely requirement for costimulatory reverse signaling through Fasl on T cells that successfully infiltrate tumors, a mechanism triggered by the induction of Fas expression on myeloid-derived cells by IL-12 and the subsequent collapse of the tumor stroma.
- Published
- 2013
- Full Text
- View/download PDF
43. Retinoic acid controls the homeostasis of pre-cDC–derived splenic and intestinal dendritic cells
- Author
-
Anthony J. Leonardi, Madhusudhanan Sukumar, Yasmine Belkaid, John R. Grainger, Christopher A. Klebanoff, Douglas C. Palmer, Sean P. Spencer, Christoph Wilhelm, Jinshan Wang, Christopher D. Scott, Gabriela A. Ferreyra, Rahul Roychoudhuri, Junfeng Sun, Joseph L. Napoli, Yun Ji, Parizad Torabi-Parizi, Nicholas P. Restifo, Pawel Muranski, Luca Gattinoni, Rongman Cai, Robert L. Danner, Jason A. Hall, and Zachary A. Borman
- Subjects
Receptors, Retinoic Acid ,Cellular differentiation ,Retinoic acid ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Neoplasms ,Immunology and Allergy ,Homeostasis ,Retinoid ,Intestinal Mucosa ,Vitamin A ,2. Zero hunger ,0303 health sciences ,hemic and immune systems ,Cell Differentiation ,3. Good health ,Intestines ,medicine.anatomical_structure ,Phenotype ,Organ Specificity ,Female ,Signal transduction ,Whole-Body Irradiation ,medicine.drug ,Signal Transduction ,medicine.drug_class ,Cell Survival ,Immunology ,Spleen ,chemical and pharmacologic phenomena ,Tretinoin ,Biology ,Article ,Immunophenotyping ,03 medical and health sciences ,Immune system ,medicine ,Animals ,Humans ,030304 developmental biology ,Cell Proliferation ,Histocompatibility Antigens Class II ,Dendritic Cells ,chemistry ,030215 immunology - Abstract
Retinoic acid is required to maintain pre-DC–derived CD11b+CD8α−Esamhigh dendritic cells (DCs) in the spleen and CD11b+CD103+ DCs in the gut., Dendritic cells (DCs) comprise distinct populations with specialized immune-regulatory functions. However, the environmental factors that determine the differentiation of these subsets remain poorly defined. Here, we report that retinoic acid (RA), a vitamin A derivative, controls the homeostasis of pre-DC (precursor of DC)–derived splenic CD11b+CD8α−Esamhigh DCs and the developmentally related CD11b+CD103+ subset within the gut. Whereas mice deprived of RA signaling significantly lost both of these populations, neither pre-DC–derived CD11b−CD8α+ and CD11b−CD103+ nor monocyte-derived CD11b+CD8α−Esamlow or CD11b+CD103− DC populations were deficient. In fate-tracking experiments, transfer of pre-DCs into RA-supplemented hosts resulted in near complete conversion of these cells into the CD11b+CD8α− subset, whereas transfer into vitamin A–deficient (VAD) hosts caused diversion to the CD11b−CD8α+ lineage. As vitamin A is an essential nutrient, we evaluated retinoid levels in mice and humans after radiation-induced mucosal injury and found this conditioning led to an acute VAD state. Consequently, radiation led to a selective loss of both RA-dependent DC subsets and impaired class II–restricted auto and antitumor immunity that could be rescued by supplemental RA. These findings establish a critical role for RA in regulating the homeostasis of pre-DC–derived DC subsets and have implications for the management of patients with immune deficiencies resulting from malnutrition and irradiation.
- Published
- 2013
44. Ionic immune suppression within the tumour microenvironment limits T cell effector function
- Author
-
Robert L. Eil, Jenny H. Pan, David S. Schrump, Douglas C. Palmer, Shashank J. Patel, Tori N. Yamamoto, Valentina Carbonaro, Klaus Okkenhaug, Nicholas P. Restifo, David Clever, Rahul Roychoudhuri, Geoffrey Guittard, Zhiya Yu, Alena Gros, Suman K. Vodnala, W. Marston Linehan, Madhusudhanan Sukumar, Christopher A. Klebanoff, Carbonaro, Valentina [0000-0003-0915-6901], Okkenhaug, Klaus [0000-0002-9432-4051], Roychoudhuri, Rahul [0000-0002-5392-1853], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Male ,Potassium Channels ,Potassium ions ,T cell ,medicine.medical_treatment ,T-Lymphocytes ,Receptors, Antigen, T-Cell ,CD8-Positive T-Lymphocytes ,Biology ,Article ,Membrane Potentials ,03 medical and health sciences ,Mice ,Necrosis ,Lymphocytes, Tumor-Infiltrating ,Immune system ,Neoplasms ,medicine ,Immune Tolerance ,Tumor Microenvironment ,Animals ,Humans ,Receptor ,Melanoma ,Multidisciplinary ,Kv1.3 Potassium Channel ,Effector ,TOR Serine-Threonine Kinases ,T-cell receptor ,Immunotherapy ,Cations, Monovalent ,Survival Analysis ,Potassium channel ,3. Good health ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Commentary ,Potassium ,Tumor Escape ,Signal transduction ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
Tumours progress despite being infiltrated by tumour-specific effector T cells1. Tumours contain areas of cellular necrosis, which is associated with poor survival in a variety of cancers2. Here, we show that necrosis releases an intracellular ion, potassium, into the extracellular fluid of mouse and human tumours causing profound suppression of T cell effector function. We find that elevations in the extracellular potassium concentration [K+]e act to impair T cell receptor (TCR)-driven Akt-mTOR phosphorylation and effector programmes, this potassium-mediated suppression of Akt-mTOR signalling and T cell function is dependent upon the activity of the serine/threonine phosphatase PP2A3,4. While the suppressive effect mediated by elevated [K+]e is independent of changes in plasma membrane potential (Vm), it does require an increase in intracellular potassium ([K+]i). Concordantly, ionic reprogramming of tumour-specific T cells through overexpression of the potassium channel Kv1.3 lowers [K+]i and improves effector functions in vitro and in vivo. Consequently, Kv1.3 T cell overexpression enhances tumour clearance and survival of melanoma-bearing mice. These results uncover a previously undescribed ionic checkpoint blocking T cell function within tumours and identify new strategies for cancer immunotherapy.
- Published
- 2016
45. Inhibition of TGF-β signaling in genetically engineered tumor antigen-reactive T cells significantly enhances tumor treatment efficacy
- Author
-
Richard A. Morgan, Steven A. Rosenberg, Nicholas P. Restifo, Zhiya Yu, Pawel Muranski, Douglas C. Palmer, and Ling Zhang
- Subjects
Cellular immunity ,Recombinant Fusion Proteins ,T-Lymphocytes ,medicine.medical_treatment ,Genetic Vectors ,Melanoma, Experimental ,Antigen-Presenting Cells ,Apoptosis ,CD8-Positive T-Lymphocytes ,Protein Serine-Threonine Kinases ,Biology ,Article ,Mice ,Cancer immunotherapy ,Transforming Growth Factor beta ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,IL-2 receptor ,Molecular Biology ,Receptor, Transforming Growth Factor-beta Type II ,Immunotherapy ,Molecular biology ,Tumor antigen ,Immunoglobulin Fc Fragments ,Retroviridae ,Treatment Outcome ,Cytokine ,Immunoglobulin G ,Cancer research ,Molecular Medicine ,Tumor promotion ,Genetic Engineering ,Receptors, Transforming Growth Factor beta ,CD8 ,Signal Transduction - Abstract
Transforming growth factor β (TGF-β) is a cytokine with complex biological functions that may involve tumor promotion or tumor suppression. It has been reported that multiple types of tumors secrete TGF-β, which can inhibit tumor-specific cellular immunity and may represent a major obstacle to the success of tumor immunotherapy. In this study, we sought to enhance tumor immunotherapy using genetically modified antigen-specific T cells by interfering with TGF-β signaling. We constructed three γ-retroviral vectors, one that expressed TGF-β-dominant-negative receptor II (DNRII) or two that secreted soluble TGF-β receptors: soluble TGF-β receptor II (sRII) and the sRII fused with mouse IgG Fc domain (sRIIFc). We demonstrated that T cells genetically modified with these viral vectors were resistant to exogenous TGF-β-induced smad-2 phosphorylation in vitro. The functionality of antigen-specific T cells engineered to resist TGF-β signaling was further evaluated in vivo using the B16 melanoma tumor model. Antigen-specific CD8+ T cells (pmel-1) or CD4+ T cells (tyrosinase-related protein-1) expressing DNRII dramatically improved tumor treatment efficacy. There was no enhancement in the B16 tumor treatment using cells secreting soluble receptors. Our data support the potential application of the blockade of TGF-β signaling in tumor-specific T cells for cancer immunotherapy.
- Published
- 2012
- Full Text
- View/download PDF
46. Repression of the DNA-binding inhibitor Id3 by Blimp-1 limits the formation of memory CD8+ T cells
- Author
-
Madhusudhanan Sukumar, Christopher A. Klebanoff, Nicholas P. Restifo, Zoltan Pos, Mahadev Rao, Robert Reger, Ena Wang, Zhiya Yu, Yun Ji, Pawel Muranski, Douglas C. Palmer, Zachary A. Borman, Luca Gattinoni, Francesco M. Marincola, and David S. Schrump
- Subjects
Regulation of gene expression ,Cell culture ,Effector ,Cellular differentiation ,Immunology ,Immunology and Allergy ,Cytotoxic T cell ,Biology ,Psychological repression ,Transcription factor ,Molecular biology ,CD8 - Abstract
The transcriptional repressor Blimp-1 promotes the differentiation of CD8(+) T cells into short-lived effector cells (SLECs) that express the lectin-like receptor KLRG-1, but how it operates remains poorly defined. Here we show that Blimp-1 bound to and repressed the promoter of the gene encoding the DNA-binding inhibitor Id3 in SLECs. Repression of Id3 by Blimp-1 was dispensable for SLEC development but limited the ability of SLECs to persist as memory cells. Enforced expression of Id3 was sufficient to restore SLEC survival and enhanced recall responses. Id3 function was mediated in part through inhibition of the transcriptional activity of E2A and induction of genes regulating genome stability. Our findings identify the Blimp-1-Id3-E2A axis as a key molecular switch that determines whether effector CD8(+) T cells are programmed to die or enter the memory pool.
- Published
- 2011
- Full Text
- View/download PDF
47. Determinants of Successful CD8+ T-Cell Adoptive Immunotherapy for Large Established Tumors in Mice
- Author
-
Christian S. Hinrichs, Steven E. Finkelstein, Sid P. Kerkar, Steven A. Rosenberg, Luca Gattinoni, Douglas C. Palmer, Pawel Muranski, Yun Ji, Christopher D. Scott, Nicholas P. Restifo, Christopher A. Klebanoff, and Zachary A. Borman
- Subjects
Interleukin 2 ,Cancer Research ,Adoptive cell transfer ,medicine.medical_treatment ,Melanoma, Experimental ,Mice, Transgenic ,CD8-Positive T-Lymphocytes ,Biology ,Cancer Vaccines ,Immunotherapy, Adoptive ,Article ,Mice ,Antigen ,Cell Line, Tumor ,medicine ,Animals ,Cytotoxic T cell ,Interleukin-15 ,Dose-Response Relationship, Drug ,Interleukin-7 ,Interleukins ,Melanoma ,Cell Differentiation ,Immunotherapy ,medicine.disease ,Tumor Burden ,Mice, Inbred C57BL ,Treatment Outcome ,Cytokine ,Oncology ,Interleukin 15 ,Immunology ,Cytokines ,Interleukin-2 ,Regression Analysis ,Immunologic Memory ,medicine.drug - Abstract
Purpose: Adoptive cell transfer (ACT) of tumor infiltrating or genetically engineered T cells can cause durable responses in patients with metastatic cancer. Multiple clinically modifiable parameters can comprise this therapy, including cell dose and phenotype, in vivo antigen restimulation, and common gamma-chain (γc) cytokine support. However, the relative contributions of each these individual components to the magnitude of the antitumor response have yet to be quantified. Experimental Design: To systematically and quantitatively appraise each of these variables, we employed the Pmel-1 mouse model treating large, established B16 melanoma tumors. In addition to cell dose and magnitude of in vivo antigen restimulation, we also evaluated the relative efficacy of central memory (TCM), effector memory (TEM), and stem cell memory (TSCM) subsets on the strength of tumor regression as well as the dose and type of clinically available γc cytokines, including IL-2, IL-7, IL-15, and IL-21. Results: We found that cell dose, T-cell differentiation status, and viral vaccine titer each were correlated strongly and significantly with the magnitude of tumor regression. Surprisingly, although the total number of IL-2 doses was correlated with tumor regression, no significant benefit to prolonged (≥6 doses) administration was observed. Moreover, the specific type and dose of γc cytokine only moderately correlated with response. Conclusion: Collectively, these findings elucidate some of the key determinants of successful ACT immunotherapy for the treatment of cancer in mice and further show that γc cytokines offer a similar ability to effectively drive antitumor T-cell function in vivo. Clin Cancer Res; 17(16); 5343–52. ©2011 AACR.
- Published
- 2011
- Full Text
- View/download PDF
48. Transplantation of mouse HSCs genetically modified to express a CD4-restricted TCR results in long-term immunity that destroys tumors and initiates spontaneous autoimmunity
- Author
-
Jon N. Marsh, Hal E. Broxmeyer, Nicholas P. Restifo, Christopher E. Touloukian, Nicholas D. Klemen, Kenneth Cornetta, Giao Hangoc, Andrew G. Brandmaier, Douglas C. Palmer, Sung P. Ha, and Garrett Kinnebrew
- Subjects
CD4-Positive T-Lymphocytes ,Genetically modified mouse ,Adoptive cell transfer ,medicine.medical_treatment ,T cell ,Receptors, Antigen, T-Cell ,Vitiligo ,Autoimmunity ,Mice, Transgenic ,Mice, SCID ,In Vitro Techniques ,Biology ,Autoimmune Diseases ,Mice ,Melanocyte differentiation ,Mice, Inbred NOD ,Transduction, Genetic ,Cell Line, Tumor ,HLA-DR4 Antigen ,medicine ,Animals ,Humans ,T-cell receptor ,Hematopoietic Stem Cell Transplantation ,Neoplasms, Experimental ,General Medicine ,Immunotherapy ,Mice, Inbred C57BL ,Transplantation ,medicine.anatomical_structure ,Immunology ,Cancer research ,CD8 ,Research Article - Abstract
The development of effective cancer immunotherapies has been consistently hampered by several factors, including an inability to instigate long-term effective functional antitumor immunity. This is particularly true for immunotherapies that focus on the adoptive transfer of activated or genetically modified mature CD8+ T cells. In this study, we sought to alter and enhance long-term host immunity by genetically modifying, then transplanting, mouse HSCs. We first cloned a previously identified tumor-reactive HLA-DR4–restricted CD4+ TCR specific for the melanocyte differentiation antigen tyrosinase-related protein 1 (Tyrp1), then constructed both a high-expression lentivirus vector and a TCR-transgenic mouse expressing the genes encoding this TCR. Using these tools, we demonstrated that both mouse and human HSCs established durable, high-efficiency TCR gene transfer following long-term transplantation into lethally irradiated mice transgenic for HLA-DR4. Recipients of genetically modified mouse HSCs developed spontaneous autoimmune vitiligo that was associated with the presence of a Th1-polarized memory effector CD4+ T cell population that expressed the Tyrp1-specific TCR. Most importantly, large numbers of CD4+ T cells expressing the Tyrp1-specific TCR were detected in secondary HLA-DR4–transgenic transplant recipients, and these mice were able to destroy subcutaneously administered melanoma cells without the aid of vaccination, immune modulation, or cytokine administration. These results demonstrate the creation of what we believe to be a novel translational model of durable lentiviral gene transfer that results in long-term effective immunity.
- Published
- 2010
- Full Text
- View/download PDF
49. Tumor-Specific CD8+ T Cells Expressing Interleukin-12 Eradicate Established Cancers in Lymphodepleted Hosts
- Author
-
Ling Zhang, Sid P. Kerkar, Douglas C. Palmer, Luis Sanchez-Perez, Zhiya Yu, Pawel Muranski, Steven A. Rosenberg, Luca Gattinoni, Andrew Kaiser, Giorgio Trinchieri, Zachary A. Borman, Robert Reger, Nicholas P. Restifo, Andrea Boni, and Richard A. Morgan
- Subjects
Cancer Research ,Adoptive cell transfer ,medicine.medical_treatment ,Melanoma, Experimental ,Mice, Transgenic ,CD8-Positive T-Lymphocytes ,Biology ,Protein Engineering ,Immunotherapy, Adoptive ,Article ,Mice ,Lymphocytes, Tumor-Infiltrating ,Transduction, Genetic ,medicine ,Animals ,Cytotoxic T cell ,Tumor microenvironment ,Interleukin ,FOXP3 ,Immunotherapy ,Flow Cytometry ,Interleukin-12 ,Mice, Inbred C57BL ,Retroviridae ,Oncology ,Immunology ,Interleukin 12 ,Female ,CD8 - Abstract
T-cell–based immunotherapies can be effective in the treatment of large vascularized tumors, but they rely on adoptive transfer of substantial numbers (∼20 million) of tumor-specific T cells administered together with vaccination and high-dose interleukin (IL)-2. In this study, we report that ∼10,000 T cells gene-engineered to express a single-chain IL-12 molecule can be therapeutically effective against established tumors in the absence of exogenous IL-2 and vaccine. Although IL-12–engineered cells did not perist long-term in hosts, they exhibited enhanced functionality and were detected in higher numbers intratumorally along with increased numbers of endogenous natural killer and CD8+ T cells just before regression. Importantly, transferred T cells isolated from tumors stably overproduced supraphysiologic amounts of IL-12, and the therapeutic effect of IL-12 produced within the tumor microenvironment could not be mimicked with high doses of exogenously provided IL-12. Furthermore, antitumor effects could be recapitulated by engineering wild-type open-repertoire splenocytes to express both the single-chain IL-12 and a recombinant tumor-specific T-cell receptor (TCR), but only when individual cells expressed both the TCR and IL-12, indicating that arrested migration of T cells at the tumor site was required for their activities. Successful tumor eradication was dependent on a lymphodepleting preconditioning regimen that reduced the number of intratumoral CD4+ Foxp3+ T regulatory cells. Our findings reveal an approach to genetically modify T cells to reduce the cell number needed, eliminate the need for vaccines or systemic IL-2, and improve immunotherapy efficacy based on adoptive transfer of gene-engineered T cells. Cancer Res; 70(17); 6725–34. ©2010 AACR.
- Published
- 2010
- Full Text
- View/download PDF
50. IFN-γ-receptor signaling ameliorates transplant vasculopathy through attenuation of CD8+T-cell-mediated injury of vascular endothelial cells
- Author
-
Matthias W. Hoffmann, Daniel S. Engeler, Yinghua Tian, Douglas C. Palmer, Sonja Firner, Burkhard Ludewig, Pierre-Alain Clavien, Simone Miller, Philippe Krebs, Nicholas P. Restifo, and Beatrice Bolinger
- Subjects
Graft Rejection ,Male ,Adoptive cell transfer ,Immunology ,Priming (immunology) ,Enzyme-Linked Immunosorbent Assay ,Mice, Transgenic ,Cell Separation ,CD8-Positive T-Lymphocytes ,Article ,Mice ,Downregulation and upregulation ,PD-L1 ,Animals ,Immunology and Allergy ,Cytotoxic T cell ,Receptor ,Receptors, Interferon ,Feedback, Physiological ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Endothelial Cells ,Flow Cytometry ,Adoptive Transfer ,Mice, Inbred C57BL ,Transplantation ,biology.protein ,Cancer research ,Heart Transplantation ,Female ,CD8 ,Signal Transduction - Abstract
Occlusive transplant vasculopathy (TV) is the major cause for chronic graft rejection. Since endothelial cells (EC) are the first graft cells encountered by activated host lymphocytes, it is important to delineate the molecular mechanisms that coordinate the interaction of EC with activated T cells. Here, the interaction of CD8(+) T cells with Ag-presenting EC in vivo was examined using a transgenic heart transplantation model with beta-galactosidase (beta-gal) expression exclusively in EC (Tie2-LacZ hearts). We found that priming with beta-gal peptide-loaded DC failed to generate a strong systemic IFN-gamma response, but elicited pronounced TV in both IFN-gamma receptor (IFNGR)-competent, and ifngr(-/-) Tie2-LacZ hearts. In contrast, stimulation of EC-specific CD8(+) T cells with beta-gal-recombinant mouse cytomegalovirus (MCMV-LacZ) in recipients of ifngr(+/+) Tie2-LacZ hearts did not precipitate significant TV. However, MCMV-LacZ infection of recipients of ifngr(-/-) Tie2-LacZ hearts led to massive activation of beta-gal-specific CD8 T cells, and led to development of fulminant TV. Further analyses revealed that the strong systemic IFN-gamma "storm" associated with MCMV infection induced upregulation of programmed death-1 ligand 1 (PD-L1) on EC, and subsequent attenuation of programmed death-1 (PD-1)-expressing EC-specific CD8(+) T cells. Thus, IFNGR signaling in ECs activates a potent peripheral negative feedback circuit that protects vascularized grafts from occlusive TV.
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.