CD20-specific adoptive immunotherapy for lymphoma using a chimeric antigen receptor with both CD28 and 4-1BB domains: pilot clinical trial results by Brian.

Slides:



Advertisements
Similar presentations
Volume 14, Issue 1, Pages (January 2012)
Advertisements

Volume 10, Issue 2, Pages (August 2004)
Supplementary Figure S1
An anti-CD20–IL-2 immunocytokine is highly efficacious in a SCID mouse model of established human B lymphoma by Stephen D. Gillies, Yan Lan, Steven Williams,
Circulating IL-15 exists as heterodimeric complex with soluble IL-15Rα in human and mouse serum by Cristina Bergamaschi, Jenifer Bear, Margherita Rosati,
William H. D. Hallett, Weiqing Jing, William R. Drobyski, Bryon D
Role of γ/δ T cells in a patient with CD4+ CD3– lymphocytosis, hypereosinophilia, and high levels of IgE  Ilan Bank, MDa,d, Avner Reshef, MDb, Miriam.
Volume 25, Issue 10, Pages (October 2017)
Volume 49, Issue 1, Pages (January 2006)
by Masih Ostad, Margareta Andersson, Astrid Gruber, and Anne Sundblad
Characterization of a humanized IgG4 anti-HLA-DR monoclonal antibody that lacks effector cell functions but retains direct antilymphoma activity and increases.
by Rafijul Bari, Teresa Bell, Wai-Hang Leung, Queenie P
TCL1: a shared tumor-associated antigen for immunotherapy against B-cell lymphomas by Jinsheng Weng, Seema Rawal, Fuliang Chu, Hyun Jun Park, Rakesh Sharma,
Lineages of human T-cell clones, including T helper 17/T helper 1 cells, isolated at different stages of anti–factor VIII immune responses by Ruth A. Ettinger,
Potential Role of a Mismatched HLA-Specific CTL Clone Developed Pre-Transplant in Graft Rejection following Cord Blood Transplantation  Hiroto Narimatsu,
Epstein-Barr virus–specific human T lymphocytes expressing antitumor chimeric T-cell receptors: potential for improved immunotherapy by Claudia Rossig,
Human NK cell development in NOD/SCID mice receiving grafts of cord blood CD34+ cells by Christian P. Kalberer, Uwe Siegler, and Aleksandra Wodnar-Filipowicz.
Synergy among receptors on resting NK cells for the activation of natural cytotoxicity and cytokine secretion by Yenan T. Bryceson, Michael E. March, Hans-Gustaf.
by Norman Nausch, Ioanna E
Ex vivo induction of multiple myeloma–specific cytotoxic T lymphocytes
Macrophages from C3-deficient mice have impaired potency to stimulate alloreactive T cells by Wuding Zhou, Hetal Patel, Ke Li, Qi Peng, Marie-Bernadette.
Rapid generation of combined CMV-specific CD4+ and CD8+ T-cell lines for adoptive transfer into recipients of allogeneic stem cell transplants by Georg.
Ex Vivo Rapamycin Generates Th1/Tc1 or Th2/Tc2 Effector T Cells With Enhanced In Vivo Function and Differential Sensitivity to Post-transplant Rapamycin.
Functional Investigation of Fas Ligand Expressions in Human Non-Small Cell Lung Cancer Cells and Its Clinical Implications  Yidan Lin, MD, PhD, Lunxu.
Extranodal dissemination of non-Hodgkin lymphoma requires CD47 and is inhibited by anti-CD47 antibody therapy by Mark P. Chao, Chad Tang, Russell K. Pachynski,
by Éric Aubin, Réal Lemieux, and Renée Bazin
Preactivation with IL-12, IL-15, and IL-18 Induces CD25 and a Functional High-Affinity IL-2 Receptor on Human Cytokine-Induced Memory-like Natural Killer.
Enhancement of the host immune responses in cutaneous T-cell lymphoma by CpG oligodeoxynucleotides and IL-15 by Maria Wysocka, Bernice M. Benoit, Sarah.
Interaction between B7-H1 and PD-1 determines initiation and reversal of T-cell anergy by Fumihiko Tsushima, Sheng Yao, Tahiro Shin, Andrew Flies, Sarah.
Simple conditioning with monospecific CD4+CD25+ regulatory T cells for bone marrow engraftment and tolerance to multiple gene products by David-Alexandre.
Immunologic potential of donor lymphocytes expressing a suicide gene for early immune reconstitution after hematopoietic T-cell–depleted stem cell transplantation.
Immunotherapy with Donor T Cells Sensitized with Overlapping Pentadecapeptides for Treatment of Persistent Cytomegalovirus Infection or Viremia  Guenther.
Cytomegalovirus-specific T cells are primed early after cord blood transplant but fail to control virus in vivo by Suzanne M. McGoldrick, Marie E. Bleakley,
by Vladia Monsurrò, Ena Wang, Yoshisha Yamano, Stephen A
Adoptive Cellular Therapy using Cells Enriched for NKG2D+CD3+CD8+T Cells after Autologous Transplantation for Myeloma  Kenneth R. Meehan, Laleh Talebian,
FLT3 ligand administration after hematopoietic cell transplantation increases circulating dendritic cell precursors that can be activated by CpG oligodeoxynucleotides.
by Bindu Varghese, Adam Widman, James Do, Behnaz Taidi, Debra K
Functionally associated targets in mantle cell lymphoma as defined by DNA microarrays and RNA interference by Eva Ortega-Paino, Johan Fransson, Sara Ek,
Cytomegalovirus-Specific Cytotoxic T Lymphocytes Can Be Efficiently Expanded from Granulocyte Colony-Stimulating Factor–Mobilized Hemopoietic Progenitor.
Virally infected and matured human dendritic cells activate natural killer cells via cooperative activity of plasma membrane-bound TNF and IL-15 by Lazar.
Volume 25, Issue 3, Pages (March 2017)
Soluble PD-1 ligands regulate T-cell function in Waldenstrom macroglobulinemia by Shahrzad Jalali, Tammy Price-Troska, Jonas Paludo, Jose Villasboas, Hyo-Jin.
Volume 18, Issue 5, Pages (May 2003)
Volume 26, Issue 2, Pages (February 2018)
Volume 17, Issue 8, Pages (August 2009)
Kathleen R. Bartemes, BA, Gail M. Kephart, BS, Stephanie J
Volume 140, Issue 4, Pages (April 2011)
Volume 26, Issue 4, Pages (April 2018)
by Kamira Maharaj, John J
Volume 24, Issue 9, Pages (September 2016)
Volume 26, Issue 4, Pages (April 2018)
Molecular Therapy - Oncolytics
Induction of T-Cell Responses against Cutaneous T-Cell Lymphomas Ex Vivo by Autologous Dendritic Cells Transfected with Amplified Tumor mRNA  Xiao Ni,
Reversal of Tumor Immune Inhibition Using a Chimeric Cytokine Receptor
Volume 24, Issue 9, Pages (September 2016)
Volume 25, Issue 9, Pages (September 2017)
Suppression of Murine Colitis and its Associated Cancer by Carcinoembryonic Antigen- Specific Regulatory T Cells  Dan Blat, Ehud Zigmond, Zoya Alteber,
CD25 expression distinguishes functionally distinct alloreactive CD4+ CD134+ (OX40+) T-cell subsets in acute graft-versus-host disease  Philip R Streeter,
Volume 23, Issue 1, Pages (January 2015)
In Vivo Expansion of Regulatory T cells With IL-2/IL-2 mAb Complexes Prevents Anti- factor VIII Immune Responses in Hemophilia A Mice Treated With Factor.
Potential Role of a Mismatched HLA-Specific CTL Clone Developed Pre-Transplant in Graft Rejection following Cord Blood Transplantation  Hiroto Narimatsu,
Oktay Kirak, MD, Gert Riethmüller, MD 
by Hakan Köksal, Pierre Dillard, Sarah E
Volume 21, Issue 11, Pages (November 2013)
Genetic Immunization With In Vivo Dendritic Cell-targeting Liposomal DNA Vaccine Carrier Induces Long-lasting Antitumor Immune Response  Arup Garu, Gopikrishna.
Dissecting the Multifactorial Causes of Immunodominance in Class I–Restricted T Cell Responses to Viruses  Weisan Chen, Luis C. Antón, Jack R. Bennink,
Adoptive transfer of gene-engineered CD4+ helper T cells induces potent primary and secondary tumor rejection by Maria Moeller, Nicole M. Haynes, Michael.
Anti-CD20 CAR mRNA enhances exPBNK in vitro cytolytic activity against CD20+ B-NHL cells and rituximab-resistant cells. exPBNK were electroporated in the.
Volume 25, Issue 9, Pages (September 2017)
T Cells with Chimeric Antigen Receptors Have Potent Antitumor Effects and Can Establish Memory in Patients with Advanced Leukemia by Michael Kalos, Bruce.
Presentation transcript:

CD20-specific adoptive immunotherapy for lymphoma using a chimeric antigen receptor with both CD28 and 4-1BB domains: pilot clinical trial results by Brian G. Till, Michael C. Jensen, Jinjuan Wang, Xiaojun Qian, Ajay K. Gopal, David G. Maloney, Catherine G. Lindgren, Yukang Lin, John M. Pagel, Lihua E. Budde, Andrew Raubitschek, Stephen J. Forman, Philip D. Greenberg, Stanley R. Riddell, and Oliver W. Press Blood Volume 119(17):3940-3950 April 26, 2012 ©2012 by American Society of Hematology

Expansion and cytotoxicity of modified T cells. Expansion and cytotoxicity of modified T cells. (A) Schematic representation of the αCD20-28-BB-ζ chimeric receptor, not to scale. (B-C) Growth curves of ex vivo expanded T cells for the 3 treated patients (B) and of the 2 attempts to expand cells from patient UPN-01 (C). PBMCs collected by apheresis were stimulated with anti-CD3 Ab and IL-2, transfected by electroporation by the plasmid encoding the CAR shown in panel A, selected with G418, and restimulated every 12 to 15 days in a rapid expansion protocol. Black bars represent periods of G418 selection; and gray bar, the target cell dose range. (D) CD20-specific cytotoxicity of G418-selected autologous patient T cells was assessed with 5-hour chromium-release assays using the following 51Cr-labeled target cells: Granta cells (MCL), Daudi cells (Burkitt NHL), EL4 cells transfected to express CD20, or the parental EL4 cell line lacking CD20 expression. The cells were tested after G418 selection (top panels) and 54, 27, and 40 days later for the 3 patients at the time of T-cell infusions (bottom panels). Data represent the mean of triplicate values, and error bars represent SEM. Brian G. Till et al. Blood 2012;119:3940-3950 ©2012 by American Society of Hematology

Immunophenotype and relative telomere length of infused T cells. Immunophenotype and relative telomere length of infused T cells. End of production T cells for patients UPN-02, UPN-03, and UPN-04, as well as cells from patient UPN-01 (after 9 stimulation cycles) were analyzed by flow cytometry after being thawed, washed, and stained with the antibodies shown. (A) Geometric mean fluorescence intensity (MFI) after subtracting the isotype control MFI and (B) percent positive cells. (C) Genomic DNA was harvested from CD3-selected apheresis PBMCs and from pre-infusion, ex vivo expanded CAR+ T cells for each patient. T cells from both attempts at expansion for UPN-01 were analyzed. Quantitative PCR was used to determine the amounts of telomeric DNA (T) and of a single-copy internal reference gene (S) for each sample. The relative mean telomere length of each cell population is represented by the T/S ratio. Data represent the mean of 2 assays, each performed in triplicate. Brian G. Till et al. Blood 2012;119:3940-3950 ©2012 by American Society of Hematology

Engraftment, persistence, and localization of modified T cells to tumor sites. Engraftment, persistence, and localization of modified T cells to tumor sites. Quantitative real-time PCR was performed on genomic DNA harvested from study subject PBMCs collected before and at serial time points after T-cell infusions, using primers specific for the transgene. CAR+ cells are shown as a percentage of total PBMCs for each of the treated patients over (A) the first month and (B) the first year. The same quantitative PCR assay was used to detect modified cells in genomic DNA harvested from surgically excised lymph nodes (C) or bone marrow aspirates (D) collected 24 to 48 hours after the third T-cell infusion. For patient UPN-03, 2 adjacent lymph nodes were excised and analyzed separately, and an additional bone marrow aspirate was performed 1 month after the third T-cell infusion. Data represent mean values (± SD) of at least 2 assays per time point, with each sample performed in triplicate. Arrows indicate infusions of genetically modified T cells; and black bar, the 14-day period of low-dose IL-2 injections. Brian G. Till et al. Blood 2012;119:3940-3950 ©2012 by American Society of Hematology

Clinical response to T-cell infusions. Clinical response to T-cell infusions. Patient UPN-04 was treated with CY 1 g/m2 2 days before the first of 3 infusions of αCD20-28-BB-ζ CAR+ T cells, which were given 2 to 5 days apart and followed by 14 days of subcutaneous IL-2 injections. (A) Response in a cervical lymph node as imaged by CT scan of the neck at baseline and 1, 6, and 12 months after T-cell infusions. (B) PET-CT scan of the cervical lymph node at baseline and 3 months after T-cell infusions. (C) Change in tumor volume over time. The sum of the products of the diameters of the 7 largest lymph nodes seen on the baseline and 1-, 6-, and 12-month CT scans. (D) Heterogeneity of response in individual lymph nodes. The products of the diameters of the 7 largest lymph nodes from the 6-month CT scan are shown as a percentage of their baseline size. L indicates left; and R, right. Brian G. Till et al. Blood 2012;119:3940-3950 ©2012 by American Society of Hematology

Effects of CY lymphodepletion and IL-2 on lymphocyte subsets and cytokine levels. Effects of CY lymphodepletion and IL-2 on lymphocyte subsets and cytokine levels. Multiparameter flow cytometry was performed with patient peripheral blood samples collected at baseline and at serial time points after T-cell infusions to quantify (A) all lymphocytes and CD4+ and CD8+ T cells, expressed as percentage change from baseline and (B) absolute numbers of Treg cells (CD4+/FoxP3+) and the ratios of Treg to CD4+ T cells and CD8+ T cells for UPN-02 (●), UPN-03 (■), and UPN-04 (▴). (C) CD19+ and CD20+ B cells, expressed as percentage change from baseline. (D) Levels of multiple cytokines were measured in patient serum samples collected at serial time points after CY administration using a Luminex microbead immunoassay and expressed as fold change from baseline (day −15, day −9, and day 0 before CY for UPN-02, UPN-03, and UPN-04, respectively). The range and baseline values (listed in parentheses sequentially for patients UPN-02, UPN-03, and UPN-04) in picograms per milliliter are as follows: IL-2: < 8 to 413 (90.9, 32.7, < 8); IL-4: < 2 to 298 (50.2, 10.6, < 2); IL-6: < 2 to 174 (58.0, 11.7, 2.73); IL-7: 4.53 to 336 (49.7, 14.9, 6.41); IL-10: < 1 to 526 (41.3, 29.4, < 1); IL-12p70: < 8 to 2011 (328, 16.3, < 8); IL-15: 1.61 to 76.4 (21.5, 4.69, 1.74); IL-17: < 15 to 412 (27.3, < 15, < 15); IL-21: < 15 to 4997 (685, 151, < 15); IFN-γ: 1.6 to 23.1 (8.18, 2.52, 1.70); TNF-α: < 3.6 to 164 (28.6, 8.58, < 3.6); and MIP1-α: < 300 to 868 (335, < 300, < 300). Arrows indicate infusions of genetically modified T cells; and black bar, the 14-day period of low-dose IL-2 injections. Brian G. Till et al. Blood 2012;119:3940-3950 ©2012 by American Society of Hematology

Immune responses to modified T cells. Immune responses to modified T cells. Two assays were performed to detect Ab against the transgene in patient serum samples collected before and at serial time points after T-cell infusions. (A) An ELISA was performed testing for the presence of Ab binding to Leu16 mouse anti–human CD20 Ab (from which the αCD20-28-BB-ζ CAR is derived). Baseline patient serum was used as a negative control. Data represent the mean ± SEM of duplicate values. (B) A flow cytometric assay was performed in which serial patient serum samples were incubated with HEK-293 cells genetically modified to express the αCD20-28-BB-ζ CAR, untransfected HEK-293 cells (negative control), or autologous EBV-LCL (positive control), followed by FITC-conjugated goat anti–human F(ab′)2 Ab. The median fluorescence intensity (MFI) for each sample at various time points at the 1:2 dilution is shown for each cell line. The black bar indicates the 14-day period of low-dose IL-2 injections. (C) The presence of cellular immune responses to infused T cells was assessed by stimulating serially collected patient PBMCs with irradiated autologous EBV-LCL modified to express the αCD20-28-BB-ζ CAR and NeoR gene products (UPN-02 and UPN-03) or irradiated autologous CAR+ T cells (UPN-04) at a 2:1 responder/stimulator ratio. After two 1-week stimulations, these PBMCs were used as effectors in 51Cr-release assays in which target cells were pre-infusion modified T cells, untransfected autologous EBV-LCL, CAR+ autologous EBV-LCL, or untransfected autologous PBMCs at E:T ratios of 25:1 (UPN-03 and UPN-04) or 12.5:1 (UPN-02). The lysis of CAR+ LCL is expressed as the difference between CAR+ and CAR− LCL for patients UPN-02 and UPN-03. The mean ± SEM of triplicate wells is shown. Brian G. Till et al. Blood 2012;119:3940-3950 ©2012 by American Society of Hematology