Miki Ando, Hiromitsu Nakauchi  Experimental Hematology 

Slides:



Advertisements
Similar presentations
T cell-mediated immunity Chapter 8
Advertisements

Asilmi 08 - T CELL DEVELOPMENT TODAY T LYMPHOCYTE DEVELOPMENT.
Lecture #10 Aims Describe T cell maturation and be able to differentiate naïve and effector T cells. Differentiate the development and functions of Th1.
1 Steven A. Feldman, Ph.D. Surgery Branch, NCI MEASUREMENT CHALLENGES FOR CAR-T BIOMANUFACTURING January 29, 2016 Adoptive Cell Therapy for the Treatment.
Activation of T Lymphocytes
Immune Receptors and Signal Transduction
Targeted therapies in hematological malignancies using therapeutic monoclonal antibodies against Eph family receptors  Sara Charmsaz, Andrew M. Scott,
Socializing Individualized T-Cell Cancer Immunotherapy
Hepatic AAV Gene Transfer and the Immune System: Friends or Foes?
CELL-MEDIATED IMMUNITY RAHUL KUMAR LOHANA 2K16/MB/50 INSTITUTE OF MICROBIOLOGY UNIVERSITY OF SINDH, JAMSHORO.
Tumor Immunity: Exploring the Role of a Checkpoint
Figure 1 CTLA-4 and PD-1–PD-L1 immune checkpoints
Chapter 9 T-cell Development
Immunotherapy of hepatocellular carcinoma
Jacques F.A.P. Miller, Michel Sadelain  Cancer Cell 
Immunological memory Topics Immune regulation  T cells
T cell mediated immunity Part II
Nat. Rev. Clin. Oncol. doi: /nrclinonc
Immunologic pathomechanism of Hodgkin's lymphoma
Figure 1 Immunomodulatory monoclonal antibodies and armoured chimeric antigen receptor (CAR) T cells overcome immune suppression Figure 1 | Immunomodulatory.
Antigen-Specific CD8 T Cells Can Eliminate Antigen-Bearing Keratinocytes with Clonogenic Potential via an IFN-γ-Dependent Mechanism  Rachel L. De Kluyver,
Antibody-modified T cells: CARs take the front seat for hematologic malignancies by Marcela V. Maus, Stephan A. Grupp, David L. Porter, and Carl H. June.
Immunologic Basis of Graft Rejection and Tolerance Following Transplantation of Liver or Other Solid Organs  Alberto Sánchez–Fueyo, Terry B. Strom  Gastroenterology 
Differentiation and Functions of CD8+ Effector T Cells
Will Engineered T Cells Expressing CD20 scFv Eradicate Melanoma?
Nat. Rev. Clin. Oncol. doi: /nrclinonc
Immunologic pathomechanism of Hodgkin's lymphoma
Colorectal cancer vaccines: Principles, results, and perspectives
Daniel Abate-Daga, Marco L Davila  Molecular Therapy - Oncolytics 
Kaitlin A. Read, Michael D. Powell, Paul W. McDonald, Kenneth J
The Many Roles of FAS Receptor Signaling in the Immune System
Volume 5, Issue 4, Pages (October 2015)
Figure 1 The role of CTLA4 and PD1 in T cell activation
Robert J Korst, MD, Ronald G Crystal, MD 
Mechanisms of immune escape in the tumor microenvironment.
Targeted therapies in hematological malignancies using therapeutic monoclonal antibodies against Eph family receptors  Sara Charmsaz, Andrew M. Scott,
Xiaoou Zhou, Malcolm K. Brenner  Experimental Hematology 
The Other Face of Chimeric Antigen Receptors
Volume 3, Issue 5, Pages (May 2003)
Ikaros: Exploiting and targeting the hematopoietic stem cell niche in B-progenitor acute lymphoblastic leukemia  Michelle L. Churchman, Charles G. Mullighan 
Turning Tumors into Vaccines: Co-opting the Innate Immune System
From pluripotent stem cells to T cells
Coinhibitory Pathways in the B7-CD28 Ligand-Receptor Family
Youry Kim, Jenny L. Anderson, Sharon R. Lewin  Cell Host & Microbe 
Figure 1 Mechanisms of action of immunotherapy modalities
Death receptor-mediated apoptosis and the liver
Tumor-Associated Macrophages: From Mechanisms to Therapy
Volume 143, Issue 1, Pages (July 2012)
Optimizing autologous cell grafts to improve stem cell gene therapy
Live and Let Die: A New Suicide Gene Therapy Moves to the Clinic
Chimeric antigen receptor T-cell therapy for solid tumors
Janna Krueger, Christopher E. Rudd  Immunity 
Cell-mediated immunity Regulation of the immune response
Peripheral Tolerance of CD8 T Lymphocytes
Cell-Intrinsic Barriers of T Cell-Based Immunotherapy
Jeffrey C Rathmell, Craig B Thompson  Cell 
Releasing the Brakes on Cancer Immunotherapy
Volume 25, Issue 4, Pages (April 2017)
Vaccines for Lung Cancer
IgA nephropathy: markers of progression and clues to pathogenesis
Dendritic-Cell-Based Therapeutic Cancer Vaccines
Plasmacytoid Dendritic Cells in Melanoma: Can We Revert Bad into Good?
Fig. 1 Engineered T cells: design of TCR versus CAR T cells.
Influence of the tumor microenvironment on FL
CAR T cell immunotherapy for human cancer
(A) Lymph node priming phase: recognition of major histocompatibility complex (MHC) by T-cell receptor (TCR), coactivating CD 28/B7 pathway activation.
Human cancer immunotherapy strategies targeting B7-H3 A, blockade of B7-H3 with blocking mAbs neutralizes inhibitory signaling in its unidentified receptor(s)
From Vaccines to Memory and Back
Memoirs of a Reincarnated T Cell
The Many Roles of FAS Receptor Signaling in the Immune System
Presentation transcript:

‘Off-the-shelf’ immunotherapy with iPSC-derived rejuvenated cytotoxic T lymphocytes  Miki Ando, Hiromitsu Nakauchi  Experimental Hematology  Volume 47, Pages 2-12 (March 2017) DOI: 10.1016/j.exphem.2016.10.009 Copyright © 2016 ISEH - International Society for Experimental Hematology Terms and Conditions

Figure 1 Immunosuppressive state of effector CD8+ T cells in the tumor microenvironment. (A) How tumors “escape” the immune system. Reduction of MHC expression diminishes presentation of target antigens. The expression of Fas ligand on tumor cells induces apoptosis in CTLs. TGFβ and IL-10 drive the differentiation of CD4+ T cells toward the regulatory phenotype, which results in T-cell inhibition. The engagement of PD-1 with its ligand programmed cell death protein ligand 1 (PD-L1) inhibits the proliferation and effector function of T cells through the recruitment of SHP2, which inactivates TCR-mediated signaling. (B) T-cell exhaustion during chronic antigen exposure. The presentation of antigenic peptides in the context of MHC class I triggers the priming of naive CD8+ T cells and their differentiation into effector CD8+ T cells. After antigen clearance, effector CD8+ T cells differentiate into memory CD8+ T cells. In contrast, during continuous exposure to antigen, T cells gradually lose effector functions and proliferation capacity and finally are eliminated via apoptosis. The gain of cell surface inhibitory receptors such as PD-1 correlated with T-cell exhaustion reduces antitumor immune function. Experimental Hematology 2017 47, 2-12DOI: (10.1016/j.exphem.2016.10.009) Copyright © 2016 ISEH - International Society for Experimental Hematology Terms and Conditions

Figure 2 The antigen recognition of T cells and CARs. (A) Antigens inside the cell are delivered to the cell surface as peptide fragments by the MHC and T cells interact with antigens through the TCR. The subsequent costimulatory signal is also necessary for optimal T-cell proliferation, differentiation, and survival. CD28 and 4-1BB interact with CD80 and 4-1BBL, respectively. (B) CARs react directly with native tumor antigens expressed on the surface of a tumor cell in a non-MHC-restricted manner. CARs are composed of antibody-binding domains fused to T-cell signaling domains. The target binding module of the single-chain antibody-variable fragment (scFv) are originated from a tumor-specific antibody. Experimental Hematology 2017 47, 2-12DOI: (10.1016/j.exphem.2016.10.009) Copyright © 2016 ISEH - International Society for Experimental Hematology Terms and Conditions

Figure 3 Rejuvenation of antigen-specific CTLs for powerful adoptive immunotherapy. Reprogramming via iPSC technology generates T-iPSCs from antigen-specific CTLs. These T-iPSCs can redifferentiate into CTLs that retain ancestral antigen specificity and cytotoxicity. Rejuvenated CTLs have higher proliferative capacity and longer telomeres than do the original exhausted CTLs. Experimental Hematology 2017 47, 2-12DOI: (10.1016/j.exphem.2016.10.009) Copyright © 2016 ISEH - International Society for Experimental Hematology Terms and Conditions

Figure 4 In vitro generation of rejuvenated antigen-specific CTLs. Antigen-specific CTL clones first are established from antigen-specific CTLs obtained from peripheral blood. These clones are reprogrammed into iPSCs (T-iPSCs) by transduction with vectors that contain the four Yamanaka reprogramming factors. For hematopoietic differentiation, T-iPSCs are cultured on C3H10T1/2 feeder cells for 2 weeks, yielding iPSC-derived sacs that contain many hematopoietic progenitor cells. Hematopoietic progenitor cells extracted from sacs are transferred onto Notch-ligand-expressing CH310T1/2 feeder cells. After 28 days of culture, T-lineage cells are harvested and stimulated, yielding huge numbers of rejuvenated antigen-specific CTLs that were generated from T-iPSCs. Experimental Hematology 2017 47, 2-12DOI: (10.1016/j.exphem.2016.10.009) Copyright © 2016 ISEH - International Society for Experimental Hematology Terms and Conditions

Figure 5 Induction of apoptosis by activating iC9. In the intrinsic cell death pathway, activated BAX or BID translocates to the mitochondria, forming pores that allow cytochrome C to move from mitochondrial intermembrane space to cytosol. There, cytochrome C activates caspase-9, which in turn activates caspase-3 and leads to apoptosis. iC9 was made by the fusion of human caspase-9 to a modified FK506-binding protein (FKBP12), allowing conditional dimerization. When CID is added, iC9 is dimerized. The dimer activates caspase-3 directly, without mitochondrial poration, inducing apoptosis. Experimental Hematology 2017 47, 2-12DOI: (10.1016/j.exphem.2016.10.009) Copyright © 2016 ISEH - International Society for Experimental Hematology Terms and Conditions

Figure 6 T-iPSCs provide a practical platform for gene therapy or engineered T-cell therapy. After reprogramming, T-iPSCs can be transduced with a suicide-gene-based safeguard system, transgenic TCRs, or CARs. Redifferentiation of T-iPSCs into which the suicide gene iC9 has been introduced yields rejCTLs that express iC9. Experimental Hematology 2017 47, 2-12DOI: (10.1016/j.exphem.2016.10.009) Copyright © 2016 ISEH - International Society for Experimental Hematology Terms and Conditions