279 results on '"Renier J, Brentjens"'
Search Results
2. Supplementary Figure Legends from Excessive Costimulation Leads to Dysfunction of Adoptively Transferred T Cells
- Author
-
Renier J. Brentjens, Sarwish Rafiq, Andrea V. Lopez, Christina Bebernitz, and Dinali Wijewarnasuriya
- Abstract
Figure Legends
- Published
- 2023
- Full Text
- View/download PDF
3. Figure S1 from Excessive Costimulation Leads to Dysfunction of Adoptively Transferred T Cells
- Author
-
Renier J. Brentjens, Sarwish Rafiq, Andrea V. Lopez, Christina Bebernitz, and Dinali Wijewarnasuriya
- Abstract
Sequences of CAR T cells with mCD8 signal peptide annotated in orange, scFv annotated in red, myc tag annotated in highlighted gray, mCD28 transmembrane and intracellular annotated in purple, mZeta annotated in green, IRES annotated in pink, mIL12 annotated in black, and m4-1BB annotated in yellow.
- Published
- 2023
- Full Text
- View/download PDF
4. Figure S2 from Excessive Costimulation Leads to Dysfunction of Adoptively Transferred T Cells
- Author
-
Renier J. Brentjens, Sarwish Rafiq, Andrea V. Lopez, Christina Bebernitz, and Dinali Wijewarnasuriya
- Abstract
(A) Media from in vitro co-culture of CAR T cells with EL4mCD19 tumor cells was assessed for IL12p70 on day 7 after CAR T cell treatment. Data shown is mean +/- s.e.m. of two independent experiments. (B) EL4mCD19 tumor bearing C57BL/6 mice, pretreated with 250mg/kg per mouse of cyclophosphamide day -3, treated i.v. with CAR T cells day 0. Significance determined bylong-rank Mantel-Cox Test, with 95% confidence interval (n=3 per group). *p=0.0224 m1928� compared to m1928� + cyclophosphamide and *p=0.0295 m19� compared to m19� + cyclophosphamide (C) Flow cytometry demonstrating CD25 expression on CAR T cells prior to injection in terms of percentage and representative flow cytometry plot (ns by ordinary one-way ANOVA). Data shown is mean +/- s.e.m. of three independent experiments (n=3 per group). (D) CAR T cells were co-cultured with EL4mCD19 tumor cells and assessed for cytotoxicity 4 hours later, using a luciferase killing assay. m1928�, m19�IL-12, and m1928�IL-12 CAR T cells showed significantly increased cytotoxicity compared to m19� CAR T cells at E:T ratios of 2:1, 1:1, and 0.5:1. There was no difference in tumor lysis between 19�12 and m1928�IL-12 CAR T cells (*p=
- Published
- 2023
- Full Text
- View/download PDF
5. Figure S3 from Excessive Costimulation Leads to Dysfunction of Adoptively Transferred T Cells
- Author
-
Renier J. Brentjens, Sarwish Rafiq, Andrea V. Lopez, Christina Bebernitz, and Dinali Wijewarnasuriya
- Abstract
(A) CAR T cells were co-cultured with EL4mCD19 tumor cells and then assessed for cytotoxicity 4 hours later, using a luciferase killing assay. There was no difference in tumor lysis between CAR T cells and vex-GFP tagged CAR T cells (two-way ANOVA). Data shown is representative of two independent experiments. (B) Flow cytometry gating strategy to determine CD8+ and CD4+ ratio and PD-1, TIM-3, and LAG-3 inhibitory receptor expression.
- Published
- 2023
- Full Text
- View/download PDF
6. Data from Optimization of T-cell Receptor–Modified T Cells for Cancer Therapy
- Author
-
Renier J. Brentjens, Christopher A. Klebanoff, Smita S. Chandran, Andrea V. Lopez, Terence J. Purdon, Sarwish Rafiq, and Dylan J. Drakes
- Abstract
T-cell receptor (TCR)–modified T-cell gene therapy can target a variety of extracellular and intracellular tumor-associated antigens, yet has had little clinical success. A potential explanation for limited antitumor efficacy is a lack of T-cell activation in vivo. We postulated that expression of proinflammatory cytokines in TCR-modified T cells would activate T cells and enhance antitumor efficacy. We demonstrate that expression of interleukin 18 (IL18) in tumor-directed TCR-modified T cells provides a superior proinflammatory signal than expression of interleukin 12 (IL12). Tumor-targeted T cells secreting IL18 promote persistent and functional effector T cells and a proinflammatory tumor microenvironment. Together, these effects augmented overall survival of mice in the pmel-1 syngeneic tumor model. When combined with sublethal irradiation, IL18-secreting pmel-1 T cells were able to eradicate tumors, whereas IL12-secreting pmel-1 T cells caused toxicity in mice through excessive cytokine secretion. In another xenograft tumor model, IL18 secretion enhanced the persistence and antitumor efficacy of NY-ESO-1–reactive TCR-modified human T cells as well as overall survival of tumor-bearing mice. These results demonstrate a rationale for optimizing the efficacy of TCR-modified T-cell cancer therapy through expression of IL18.See related commentary by Wijewarnasuriya et al., p. 732
- Published
- 2023
- Full Text
- View/download PDF
7. Figure S4 from Excessive Costimulation Leads to Dysfunction of Adoptively Transferred T Cells
- Author
-
Renier J. Brentjens, Sarwish Rafiq, Andrea V. Lopez, Christina Bebernitz, and Dinali Wijewarnasuriya
- Abstract
(A) CAR T cells, day 5 after infusion, were characterized by flow cytometry in terms of percentage and representative plots. m1928�IL-12 CAR T cells expressed higher levels of inhibitory receptors, PD-1 (*p=0.0015) and LAG-3 (*p=0.0491) and showed a trend toward higher levels of TIM-3 (*p=0.1715). Data obtained is mean +/- s.e.m. of three independent experiments analyzed by unpaired t test. (B) Representative flow cytometry plots of PD-1, LAG-3, and TIM-3 inhibitory receptor expression of CAR T cells referenced in Figure 2B.
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Figure Legends from Optimization of T-cell Receptor–Modified T Cells for Cancer Therapy
- Author
-
Renier J. Brentjens, Christopher A. Klebanoff, Smita S. Chandran, Andrea V. Lopez, Terence J. Purdon, Sarwish Rafiq, and Dylan J. Drakes
- Abstract
Supplementary Data Figure Legends
- Published
- 2023
- Full Text
- View/download PDF
9. Data from BCMA-Targeted CAR T-cell Therapy plus Radiotherapy for the Treatment of Refractory Myeloma Reveals Potential Synergy
- Author
-
Renier J. Brentjens, Ola Landgren, Isabelle Riviere, Andreas Rimner, Jonathan Landa, Bianca D. Santomasso, Elena Mead, Aaron D. Goldberg, Mark B. Geyer, Anthony F. Daniyan, Terence J. Purdon, Brigitte Senechal, Xiuyan Wang, Mette Staehr, Sham Mailankody, and Eric L. Smith
- Abstract
We present a case of a patient with multiply relapsed, refractory myeloma whose clinical course showed evidence of a synergistic abscopal-like response to chimeric antigen receptor (CAR) T-cell therapy and localized radiotherapy (XRT). Shortly after receiving B-cell maturation antigen (BCMA)–targeted CAR T-cell therapy, the patient required urgent high-dose steroids and XRT for spinal cord compression. Despite the steroids, the patient had a durable systemic response that could not be attributed to XRT alone. Post-XRT findings included a second wave of fever and increased CRP and IL6, beginning 21 days after CAR T cells, which is late for cytokine-release syndrome from CAR T-cell therapy alone on this trial. Given this response, which resembled cytokine-release syndrome, immediately following XRT, we investigated changes in the patient's T-cell receptor (TCR) repertoire over 10 serial time points. Comparing T-cell diversity via Morisita's overlap indices (CD), we discovered that, although the diversity was initially stable after CAR T-cell therapy compared with baseline (CD = 0.89–0.97, baseline vs. 4 time points after CAR T cells), T-cell diversity changed after the conclusion of XRT, with >30% newly expanded TCRs (CD = 0.56–0.69, baseline vs. 4 time points after XRT). These findings suggest potential synergy between radiation and CAR T-cell therapies resulting in an abscopal-like response.
- Published
- 2023
- Full Text
- View/download PDF
10. Figure S6 from Excessive Costimulation Leads to Dysfunction of Adoptively Transferred T Cells
- Author
-
Renier J. Brentjens, Sarwish Rafiq, Andrea V. Lopez, Christina Bebernitz, and Dinali Wijewarnasuriya
- Abstract
(A) B cells were assessed for CD80 expression by flow cytometry through peripheral blood. *p=0.048 unpaired t test of BM and *p=0.0233 unpaired t test of spleen. Data shown is mean +/- s.e.m. of two independent experiments. (B) Luminex data showing IL-4 secreting from EL4mCD19 tumor cells. Data shown is mean +/- s.e.m. of two independent experiments. *p=0.002 by unpaired t test. (C) Flow cytometry demonstrating lack of CD28 expression on CD28-/- T cells. Data shown is representative of two independent experiments. (D) CAR T cells were co-cultured with EL4mCD19 tumor cells and then assessed for cytotoxicity 4 hours later, using a luciferase killing assay. There was no difference in tumor lysis between CD28-/- or wildtype transduced CAR T cells. Data shown is mean +/- s.e.m. of two independent experiments.
- Published
- 2023
- Full Text
- View/download PDF
11. Data from Excessive Costimulation Leads to Dysfunction of Adoptively Transferred T Cells
- Author
-
Renier J. Brentjens, Sarwish Rafiq, Andrea V. Lopez, Christina Bebernitz, and Dinali Wijewarnasuriya
- Abstract
Although clinical responses with CD19-targeting chimeric antigen receptor (CAR) T-cell treatment have been observed in patients with certain hematologic malignancies, high rates of disease relapse highlight the necessity to understand and improve mechanisms of CAR T-cell failure. Because T-cell dysfunction is thought to contribute to CAR T-cell treatment failure, understanding what mechanisms drive T cells into this dysfunctional state may aid optimal design of efficacious CAR T cells. Dysfunctional CAR T cells have been characterized as having upregulated inhibitory receptors and decreased cytolytic capabilities. Previous studies have identified a role for sustained CAR CD3ζ signaling in CAR T-cell dysfunction. Here, we demonstrate a mechanism that drives dysfunction in CAR T cells through excessive costimulation. Fully activated CD19-targeted CAR T cells were rendered dysfunctional upon stimulation with both endogenous CD28 stimulation and CAR-mediated CD28 costimulation. Costimulation-driven dysfunction of CAR T cells was demonstrated in a syngeneic immunocompetent mouse model, in which CAR T cells were activated with signals 1 (CD3ζ), 2 (CD28), and 3 (IL12). Thus, we show that CAR T-cell dysfunction can be driven through excessive CD28 and 4-1BB costimulation.See related article by Drakes et al., p. 743
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Data from BCMA-Targeted CAR T-cell Therapy plus Radiotherapy for the Treatment of Refractory Myeloma Reveals Potential Synergy
- Author
-
Renier J. Brentjens, Ola Landgren, Isabelle Riviere, Andreas Rimner, Jonathan Landa, Bianca D. Santomasso, Elena Mead, Aaron D. Goldberg, Mark B. Geyer, Anthony F. Daniyan, Terence J. Purdon, Brigitte Senechal, Xiuyan Wang, Mette Staehr, Sham Mailankody, and Eric L. Smith
- Abstract
Supplementary Data Combined
- Published
- 2023
- Full Text
- View/download PDF
13. Figure S5 from Excessive Costimulation Leads to Dysfunction of Adoptively Transferred T Cells
- Author
-
Renier J. Brentjens, Sarwish Rafiq, Andrea V. Lopez, Christina Bebernitz, and Dinali Wijewarnasuriya
- Abstract
(A) CAR T cells, day 0 before infusion, were characterized by flow cytometry. Data shown is mean +/- s.e.m. of three independent experiments. "ns" (not significant) by unpaired t test. (B) Representative flow cytometry plots of CAR T cell CD8+ and CD4+ ratios day 5 after infusion referenced in Figure 2D. (C) Representative flow cytometry plots of PD-1, LAG-3, and TIM-3 inhibitory receptor expression of CD8+ CAR T cells referenced in Figure 2E. (D) CAR T cells, day 5 after infusion, were characterized by flow cytometry. m1928�IL-12 CAR T cells, gated on CD8+ T cells, expressed higher levels of double positive inhibitory receptors, LAG-3;TIM-3 (*p=0.0397) and LAG-3;PD-1 (*p=0.001), and showed a trend toward higher levels of TIM-3;PD-1 (*p=0.0563). Data obtained is mean +/- s.e.m. of three independent experiments analyzed by unpaired t test.
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Figures 1-8 from Optimization of T-cell Receptor–Modified T Cells for Cancer Therapy
- Author
-
Renier J. Brentjens, Christopher A. Klebanoff, Smita S. Chandran, Andrea V. Lopez, Terence J. Purdon, Sarwish Rafiq, and Dylan J. Drakes
- Abstract
Supplementary Figures 1-8
- Published
- 2023
- Full Text
- View/download PDF
15. Table S1, Table S2, Table S3, Table S4 from Clinical and Biological Correlates of Neurotoxicity Associated with CAR T-cell Therapy in Patients with B-cell Acute Lymphoblastic Leukemia
- Author
-
Renier J. Brentjens, Michel Sadelain, Hans-Guido Wendel, Mithat Gonen, Yvette Bernal, Daniel Li, Lisa M. DeAngelis, Xi Chen, Behroze Vachha, Hui Liu, Justin R. Cross, Terence Purdon, Brigitte Senechal, Xiuyan Wang, Elizabeth Halton, Elena Mead, Jessica Flynn, Isabelle Riviere, Darin Salloum, Jae H. Park, and Bianca D. Santomasso
- Abstract
Table S1: Detailed descriptions of all grade neurotoxicity; Table S2: Correlation between CAR T cell phenotype and neurotoxicity; Table S3: Correlation between CSF cytokines and CSF protein concentrations; Table S4: Definitions of CRS
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary Tables from A Phase I Trial of Regional Mesothelin-Targeted CAR T-cell Therapy in Patients with Malignant Pleural Disease, in Combination with the Anti–PD-1 Agent Pembrolizumab
- Author
-
Michel Sadelain, David R. Jones, Renier J. Brentjens, Charles M. Rudin, Mithat Gönen, William D. Travis, Xiuyan Wang, Brigitte Senechal, Devanjan Sikder, Shanu Modi, Jennifer L. Sauter, Claudia Diamonte, Alain Vincent, Erin McGee, Daniel Ngai, Jose A. Araujo Filho, Bobby Daly, Kay See Tan, Rocio Perez-Johnston, John Pineda, Elizabeth Halton, Navin K. Chintala, Waseem Cheema, Amy Zhu, Roisin E. O'Cearbhaill, Valerie W. Rusch, Stephen B. Solomon, Isabelle Rivière, Marjorie G. Zauderer, and Prasad S. Adusumilli
- Abstract
Supplemental Tables
- Published
- 2023
- Full Text
- View/download PDF
17. Data from A Phase I Trial of Regional Mesothelin-Targeted CAR T-cell Therapy in Patients with Malignant Pleural Disease, in Combination with the Anti–PD-1 Agent Pembrolizumab
- Author
-
Michel Sadelain, David R. Jones, Renier J. Brentjens, Charles M. Rudin, Mithat Gönen, William D. Travis, Xiuyan Wang, Brigitte Senechal, Devanjan Sikder, Shanu Modi, Jennifer L. Sauter, Claudia Diamonte, Alain Vincent, Erin McGee, Daniel Ngai, Jose A. Araujo Filho, Bobby Daly, Kay See Tan, Rocio Perez-Johnston, John Pineda, Elizabeth Halton, Navin K. Chintala, Waseem Cheema, Amy Zhu, Roisin E. O'Cearbhaill, Valerie W. Rusch, Stephen B. Solomon, Isabelle Rivière, Marjorie G. Zauderer, and Prasad S. Adusumilli
- Abstract
Malignant pleural diseases, comprising metastatic lung and breast cancers and malignant pleural mesothelioma (MPM), are aggressive solid tumors with poor therapeutic response. We developed and conducted a first-in-human, phase I study of regionally delivered, autologous, mesothelin-targeted chimeric antigen receptor (CAR) T-cell therapy. Intrapleural administration of 0.3M to 60M CAR T cells/kg in 27 patients (25 with MPM) was safe and well tolerated. CAR T cells were detected in peripheral blood for >100 days in 39% of patients. Following our demonstration that PD-1 blockade enhances CAR T-cell function in mice, 18 patients with MPM also received pembrolizumab safely. Among those patients, median overall survival from CAR T-cell infusion was 23.9 months (1-year overall survival, 83%). Stable disease was sustained for ≥6 months in 8 patients; 2 exhibited complete metabolic response on PET scan. Combination immunotherapy with CAR T cells and PD-1 blockade agents should be further evaluated in patients with solid tumors.Significance:Regional delivery of mesothelin-targeted CAR T-cell therapy followed by pembrolizumab administration is feasible, safe, and demonstrates evidence of antitumor efficacy in patients with malignant pleural diseases. Our data support the investigation of combination immunotherapy with CAR T cells and PD-1 blockade agents in solid tumors.See related commentary by Aldea et al., p. 2674.This article is highlighted in the In This Issue feature, p. 2659
- Published
- 2023
- Full Text
- View/download PDF
18. Figure S1, Figure S2, Figure S3, Figure S4 from Clinical and Biological Correlates of Neurotoxicity Associated with CAR T-cell Therapy in Patients with B-cell Acute Lymphoblastic Leukemia
- Author
-
Renier J. Brentjens, Michel Sadelain, Hans-Guido Wendel, Mithat Gonen, Yvette Bernal, Daniel Li, Lisa M. DeAngelis, Xi Chen, Behroze Vachha, Hui Liu, Justin R. Cross, Terence Purdon, Brigitte Senechal, Xiuyan Wang, Elizabeth Halton, Elena Mead, Jessica Flynn, Isabelle Riviere, Darin Salloum, Jae H. Park, and Bianca D. Santomasso
- Abstract
Figure S1: Detailed time course of neurotoxicity for each patient; Figure S2: Serum cytokines in relation to tocilizumab and corticosteroid administration; Figure S3: Hematopoietic toxicity and coagulopathy in severe neurotoxicity; Figure S4: Angiopoietin 1 and 2 alterations in severe neurotoxicity
- Published
- 2023
- Full Text
- View/download PDF
19. Supplementary Figures from A Phase I Trial of Regional Mesothelin-Targeted CAR T-cell Therapy in Patients with Malignant Pleural Disease, in Combination with the Anti–PD-1 Agent Pembrolizumab
- Author
-
Michel Sadelain, David R. Jones, Renier J. Brentjens, Charles M. Rudin, Mithat Gönen, William D. Travis, Xiuyan Wang, Brigitte Senechal, Devanjan Sikder, Shanu Modi, Jennifer L. Sauter, Claudia Diamonte, Alain Vincent, Erin McGee, Daniel Ngai, Jose A. Araujo Filho, Bobby Daly, Kay See Tan, Rocio Perez-Johnston, John Pineda, Elizabeth Halton, Navin K. Chintala, Waseem Cheema, Amy Zhu, Roisin E. O'Cearbhaill, Valerie W. Rusch, Stephen B. Solomon, Isabelle Rivière, Marjorie G. Zauderer, and Prasad S. Adusumilli
- Abstract
Supplemental Figures
- Published
- 2023
- Full Text
- View/download PDF
20. Supplementary Material from A Phase I Trial of Regional Mesothelin-Targeted CAR T-cell Therapy in Patients with Malignant Pleural Disease, in Combination with the Anti–PD-1 Agent Pembrolizumab
- Author
-
Michel Sadelain, David R. Jones, Renier J. Brentjens, Charles M. Rudin, Mithat Gönen, William D. Travis, Xiuyan Wang, Brigitte Senechal, Devanjan Sikder, Shanu Modi, Jennifer L. Sauter, Claudia Diamonte, Alain Vincent, Erin McGee, Daniel Ngai, Jose A. Araujo Filho, Bobby Daly, Kay See Tan, Rocio Perez-Johnston, John Pineda, Elizabeth Halton, Navin K. Chintala, Waseem Cheema, Amy Zhu, Roisin E. O'Cearbhaill, Valerie W. Rusch, Stephen B. Solomon, Isabelle Rivière, Marjorie G. Zauderer, and Prasad S. Adusumilli
- Abstract
Supplemental Material
- Published
- 2023
- Full Text
- View/download PDF
21. Multipurposing CARs: Same engine, different vehicles
- Author
-
A.K.M. Nawshad Hossian, Christopher S. Hackett, Renier J. Brentjens, and Sarwish Rafiq
- Subjects
Pharmacology ,Receptors, Chimeric Antigen ,Neoplasms ,T-Lymphocytes ,Drug Discovery ,Receptors, Antigen, T-Cell ,Genetics ,Humans ,Molecular Medicine ,Genetic Engineering ,Immunotherapy, Adoptive ,Molecular Biology - Abstract
T cells genetically engineered to recognize and eliminate tumor cells through synthetic chimeric antigen receptors (CARs) have demonstrated remarkable clinical efficacy against B cell leukemia over the past decade. This therapy is a form of highly personalized medicine that involves genetically modifying a patient's T cells to recognize and kill cancer cells. With the FDA approval of 5 CAR T cell products, this approach has been validated as a powerful new drug in the therapeutic armamentarium against cancer. Researchers are now studying how to expand this technology beyond its use in conventional polyclonal αβ T cells to address limitations to the current therapy in cancer and applications beyond it. Considering the specific characteristics of immune cell from diverse lineages, several preclinical and clinical studies are under way to assess the advantages of CAR-redirected function in these cells and apply the lessons learned from CAR T cell therapy in cancer to other diseases.
- Published
- 2022
- Full Text
- View/download PDF
22. Preparing for CAR T cell therapy: patient selection, bridging therapies and lymphodepletion
- Author
-
Leila Amini, Sara K. Silbert, Shannon L. Maude, Loretta J. Nastoupil, Carlos A. Ramos, Renier J. Brentjens, Craig S. Sauter, Nirali N. Shah, and Mohamed Abou-el-Enein
- Subjects
Oncology - Published
- 2022
- Full Text
- View/download PDF
23. Data from Successful Eradication of Established Peritoneal Ovarian Tumors in SCID-Beige Mice following Adoptive Transfer of T Cells Genetically Targeted to the MUC16 Antigen
- Author
-
Renier J. Brentjens, David R. Spriggs, Douglas A. Levine, Kay J. Park, Yan Nikhamin, Thapi D. Rao, and Alena A. Chekmasova
- Abstract
Purpose: Most patients diagnosed with ovarian cancer will ultimately die from their disease. For this reason, novel approaches to the treatment of this malignancy are needed. Adoptive transfer of a patient's own T cells, genetically modified ex vivo through the introduction of a gene encoding a chimeric antigen receptor (CAR) targeted to a tumor-associated antigen, is a novel approach to the treatment of ovarian cancer.Experimental Design: We have generated several CARs targeted to the retained extracellular domain of MUC16, termed MUC-CD, an antigen expressed on most ovarian carcinomas. We investigate the in vitro biology of human T cells retrovirally transduced to express these CARs by coculture assays on artificial antigen-presenting cells as well as by cytotoxicity and cytokine release assays using the human MUC-CD+ ovarian tumor cell lines and primary patient tumor cells. Further, we assess the in vivo antitumor efficacy of MUC-CD–targeted T cells in SCID-Beige mice bearing peritoneal human MUC-CD+ tumor cell lines.Results: CAR-modified, MUC-CD–targeted T cells exhibited efficient MUC-CD–specific cytolytic activity against both human ovarian cell and primary ovarian carcinoma cells in vitro. Furthermore, expanded MUC-CD–targeted T cells infused through either i.p. injection or i.v. infusion into SCID-Beige mice bearing orthotopic human MUC-CD+ ovarian carcinoma tumors either delayed progression or fully eradicated disease.Conclusion: These promising preclinical studies justify further investigation of MUC-CD–targeted T cells as a potential therapeutic approach for patients with high-risk MUC16+ ovarian carcinomas. Clin Cancer Res; 16(14); 3594–606. ©2010 AACR.
- Published
- 2023
- Full Text
- View/download PDF
24. Supplementary Data from Successful Eradication of Established Peritoneal Ovarian Tumors in SCID-Beige Mice following Adoptive Transfer of T Cells Genetically Targeted to the MUC16 Antigen
- Author
-
Renier J. Brentjens, David R. Spriggs, Douglas A. Levine, Kay J. Park, Yan Nikhamin, Thapi D. Rao, and Alena A. Chekmasova
- Abstract
Supplementary Figures S1-S6.
- Published
- 2023
- Full Text
- View/download PDF
25. CAR T cell therapy: looking back and looking forward
- Author
-
Marco L, Davila and Renier J, Brentjens
- Subjects
Immunotherapy, Adoptive - Published
- 2022
26. GPRC5D-Targeted CAR T Cells for Myeloma. Reply
- Author
-
Sham, Mailankody, Renier J, Brentjens, and Eric L, Smith
- Subjects
Receptors, Chimeric Antigen ,T-Lymphocytes ,Humans ,Multiple Myeloma ,Immunotherapy, Adoptive ,Receptors, G-Protein-Coupled - Published
- 2022
27. Author Correction: Gut microbiome correlates of response and toxicity following anti-CD19 CAR T cell therapy
- Author
-
Melody Smith, Anqi Dai, Guido Ghilardi, Kimberly V. Amelsberg, Sean M. Devlin, Raymone Pajarillo, John B. Slingerland, Silvia Beghi, Pamela S. Herrera, Paul Giardina, Annelie Clurman, Emmanuel Dwomoh, Gabriel Armijo, Antonio L. C. Gomes, Eric R. Littmann, Jonas Schluter, Emily Fontana, Ying Taur, Jae H. Park, Maria Lia Palomba, Elizabeth Halton, Josel Ruiz, Tania Jain, Martina Pennisi, Aishat Olaide Afuye, Miguel-Angel Perales, Craig W. Freyer, Alfred Garfall, Shannon Gier, Sunita Nasta, Daniel Landsburg, James Gerson, Jakub Svoboda, Justin Cross, Elise A. Chong, Sergio Giralt, Saar I. Gill, Isabelle Riviere, David L. Porter, Stephen J. Schuster, Michel Sadelain, Noelle Frey, Renier J. Brentjens, Carl H. June, Eric G. Pamer, Jonathan U. Peled, Andrea Facciabene, Marcel R. M. van den Brink, and Marco Ruella
- Subjects
General Medicine ,General Biochemistry, Genetics and Molecular Biology - Published
- 2022
- Full Text
- View/download PDF
28. Depletion of high-content CD14+ cells from apheresis products is critical for successful transduction and expansion of CAR T cells during large-scale cGMP manufacturing
- Author
-
Eric L. Smith, Brigitte Senechal, Craig S. Sauter, Jolanta Stefanski, Fang Du, Prasad S. Adusumilli, Susan F. Slovin, Ling-bo Shen, Devanjan S. Sikder, Jagrutiben Chaudhari, Kevin J. Curran, Renier J. Brentjens, Keyur Thummar, Yongzeng Wang, Melanie Hall, Xiuyan Wang, Isabelle Riviere, Roisin E. O'Cearbhaill, Mark B. Geyer, Sham Mailankhody, Mingzhu Zhu, Jae H. Park, Paridhi Gautam, Jinrong Qu, and Oriana Borquez-Ojeda
- Subjects
CD3 ,Cell ,Phases of clinical research ,monocyte depletion ,Pharmacology ,QH426-470 ,CD19 ,medicine ,clinical grade ,Genetics ,large-scale manufacturing ,Molecular Biology ,Multiple myeloma ,B cell ,biology ,QH573-671 ,business.industry ,Monocyte ,medicine.disease ,Chimeric antigen receptor ,cGMP ,medicine.anatomical_structure ,CAR T cell ,Immunology ,biology.protein ,Molecular Medicine ,business ,Cytology - Abstract
With the US Food and Drug Administration (FDA) approval of four CD19- and one BCMA-targeted chimeric antigen receptor (CAR) therapy for B cell malignancies, CAR T cell therapy has finally reached the status of a medicinal product. The successful manufacturing of autologous CAR T cell products is a key requirement for this promising treatment modality. By analyzing the composition of 214 apheresis products from 210 subjects across eight disease indications, we found that high CD14+ cell content poses a challenge for manufacturing CAR T cells, especially in patients with non-Hodgkin's lymphoma and multiple myeloma caused by the non-specific phagocytosis of the magnetic beads used to activate CD3+ T cells. We demonstrated that monocyte depletion via rapid plastic surface adhesion significantly reduces the CD14+ monocyte content in the apheresis products and simultaneously boosts the CD3+ content. We established a 40% CD14+ threshold for the stratification of apheresis products across nine clinical trials and demonstrated the effectiveness of this procedure by comparing manufacturing runs in two phase 1 clinical trials. Our study suggests that CD14+ content should be monitored in apheresis products, and that the manufacturing of CAR T cells should incorporate a step that lessens the CD14+ cell content in apheresis products containing more than 40% to maximize the production success.
- Published
- 2021
29. A Phase I Trial of Regional Mesothelin-Targeted CAR T-cell Therapy in Patients with Malignant Pleural Disease, in Combination with the Anti–PD-1 Agent Pembrolizumab
- Author
-
David R. Jones, Shanu Modi, Alain Vincent, William D. Travis, Marjorie G. Zauderer, Prasad S. Adusumilli, Valerie W. Rusch, Bobby Daly, Brigitte Senechal, Devanjan S. Sikder, Daniel Ngai, Jennifer L. Sauter, Waseem Cheema, Michel Sadelain, Rocio Perez-Johnston, Claudia Diamonte, Renier J. Brentjens, Jose A. Araujo Filho, Stephen B. Solomon, Amy Zhu, Elizabeth Halton, John Pineda, Xiuyan Wang, Roisin E. O'Cearbhaill, Navin K. Chintala, Kay See Tan, Erin McGee, Charles M. Rudin, Mithat Gonen, and Isabelle Riviere
- Subjects
Oncology ,medicine.medical_specialty ,Lung ,biology ,business.industry ,Anti pd 1 ,Pembrolizumab ,medicine.disease ,Chimeric antigen receptor ,Blockade ,Pleural disease ,medicine.anatomical_structure ,Internal medicine ,medicine ,biology.protein ,Mesothelin ,In patient ,business - Abstract
Malignant pleural diseases, comprising metastatic lung and breast cancers and malignant pleural mesothelioma (MPM), are aggressive solid tumors with poor therapeutic response. We developed and conducted a first-in-human, phase I study of regionally delivered, autologous, mesothelin-targeted chimeric antigen receptor (CAR) T-cell therapy. Intrapleural administration of 0.3M to 60M CAR T cells/kg in 27 patients (25 with MPM) was safe and well tolerated. CAR T cells were detected in peripheral blood for >100 days in 39% of patients. Following our demonstration that PD-1 blockade enhances CAR T-cell function in mice, 18 patients with MPM also received pembrolizumab safely. Among those patients, median overall survival from CAR T-cell infusion was 23.9 months (1-year overall survival, 83%). Stable disease was sustained for ≥6 months in 8 patients; 2 exhibited complete metabolic response on PET scan. Combination immunotherapy with CAR T cells and PD-1 blockade agents should be further evaluated in patients with solid tumors. Significance: Regional delivery of mesothelin-targeted CAR T-cell therapy followed by pembrolizumab administration is feasible, safe, and demonstrates evidence of antitumor efficacy in patients with malignant pleural diseases. Our data support the investigation of combination immunotherapy with CAR T cells and PD-1 blockade agents in solid tumors. See related commentary by Aldea et al., p. 2674. This article is highlighted in the In This Issue feature, p. 2659
- Published
- 2021
- Full Text
- View/download PDF
30. Interventions and outcomes of adult patients with B-ALL progressing after CD19 chimeric antigen receptor T-cell therapy
- Author
-
Brigitte Senechal, Mark B. Geyer, Marco L. Davila, Kevin J. Curran, Mithat Gonen, Mikhail Roshal, Isabelle Riviere, Michel Sadelain, Peter Maslak, Jessica Flynn, Kitsada Wudhikarn, Claudia Diamonte, Renier J. Brentjens, Jae H. Park, Xiuyan Wang, and Elizabeth Halton
- Subjects
Adult ,Male ,Oncology ,medicine.medical_specialty ,Clinical Trials and Observations ,Immunology ,Psychological intervention ,Immunotherapy, Adoptive ,Biochemistry ,Disease-Free Survival ,CD19 ,Refractory ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,Internal medicine ,Antibodies, Bispecific ,medicine ,Humans ,Inotuzumab Ozogamicin ,Aged ,Salvage Therapy ,Response rate (survey) ,biology ,business.industry ,Cell Biology ,Hematology ,Middle Aged ,Chimeric antigen receptor ,Survival Rate ,Natural history ,biology.protein ,Female ,Chimeric Antigen Receptor T-Cell Therapy ,Blinatumomab ,business ,human activities ,medicine.drug - Abstract
CD19-targeted chimeric antigen receptor (CAR) T-cell therapy has become a breakthrough treatment of patients with relapsed/refractory B-cell acute lymphoblastic leukemia (B-ALL). However, despite the high initial response rate, the majority of adult patients with B-ALL progress after CD19 CAR T-cell therapy. Data on the natural history, management, and outcome of adult B-ALL progressing after CD19 CAR T cells have not been described in detail. Herein, we report comprehensive data of 38 adult patients with B-ALL who progressed after CD19 CAR T therapy at our institution. The median time to progression after CAR T-cell therapy was 5.5 months. Median survival after post–CAR T progression was 7.5 months. A high disease burden at the time of CAR T-cell infusion was significantly associated with risk of post–CAR T progression. Thirty patients (79%) received salvage treatment of post–CAR T disease progression, and 13 patients (43%) achieved complete remission (CR), but remission duration was short. Notably, 7 (58.3%) of 12 patients achieved CR after blinatumomab and/or inotuzumab administered following post–CAR T failure. Multivariate analysis revealed that a longer remission duration from CAR T cells was associated with superior survival after progression following CAR T-cell therapy. In summary, overall prognosis of adult B-ALL patients progressing after CD19 CAR T cells was poor, although a subset of patients achieved sustained remissions to salvage treatments, including blinatumomab, inotuzumab, and reinfusion of CAR T cells. Novel therapeutic strategies are needed to reduce risk of progression after CAR T-cell therapy and improve outcomes of these patients.
- Published
- 2021
- Full Text
- View/download PDF
31. CAR T cell therapy: looking back and looking forward
- Author
-
Marco L. Davila and Renier J. Brentjens
- Subjects
Cancer Research ,Oncology - Published
- 2022
- Full Text
- View/download PDF
32. Defining an Optimal Dual-Targeted CAR T-cell Therapy Approach Simultaneously Targeting BCMA and GPRC5D to Prevent BCMA Escape–Driven Relapse in Multiple Myeloma
- Author
-
Eric L. Smith, Yunxin Chen, Renier J. Brentjens, Carlos Fernández de Larrea, Terence J. Purdon, Hans-Guido Wendel, Vladimir Ponomarev, Andrea V. Lopez, Khong Y. Ng, William D. Godfrey, and Mette Staehr
- Subjects
business.industry ,medicine.medical_treatment ,Cell ,General Medicine ,Immunotherapy ,medicine.disease ,Immunotherapy, Adoptive ,Chimeric antigen receptor ,Receptors, G-Protein-Coupled ,Cell therapy ,medicine.anatomical_structure ,Antigen ,medicine ,Cancer research ,Humans ,Vector (molecular biology) ,B-Cell Maturation Antigen ,Neoplasm Recurrence, Local ,Multiple Myeloma ,Receptor ,business ,Multiple myeloma - Abstract
Chimeric antigen receptor (CAR) T-cell therapy for multiple myeloma targeting B-cell maturation antigen (TNFRSF17; BCMA) induces high overall response rates; however, relapse occurs commonly. A reservoir of multiple myeloma cells lacking sufficient BCMA surface expression (antigen escape) may be implicated in relapse. We demonstrate that simultaneous targeting of an additional antigen—here, G protein-coupled receptor class-C group-5 member-D (GPRC5D)—can prevent BCMA escape–mediated relapse in a model of multiple myeloma. To identify an optimal approach, we compare subtherapeutic doses of different forms of dual-targeted cellular therapy. These include; (i) parallel-produced and pooled mono-targeted CAR T cells, (ii) bicistronic constructs expressing distinct CARs from a single vector, and (iii) a dual-scFv “single-stalk” CAR design. When targeting BCMA-negative disease, bicistronic and pooled approaches had the highest efficacy, whereas for dual-antigen–expressing disease, the bicistronic approach was more efficacious than the pooled approach. Mechanistically, expressing two CARs on a single cell enhanced the strength of CAR T-cell/target cell interactions. Significance: Myeloma frequently relapses post-CAR T-cell therapy; antigen escape–mediated relapse can be mitigated with upfront dual-targeting (BCMA/GPRC5D). A bicistronic vector encoding two CARs avoids the challenge of parallel manufacturing separate CAR T-cell products, while providing superior efficacy; this dual-targeted approach may enhance the durability of responses to cellular therapy for myeloma. See related commentary by Simon and Riddell, p. 130. This article is highlighted in the In This Issue feature, p. 127
- Published
- 2020
- Full Text
- View/download PDF
33. Cytokine IL-36γ Improves CAR T Cell Functionality and Induces Endogenous Anti-Tumor Response
- Author
-
Renier J. Brentjens, Terence J. Purdon, Anthony F. Daniyan, Xinghuo Li, and Andrea V. Lopez
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,Receptors, Antigen, T-Cell ,Antigen-Presenting Cells ,Apoptosis ,Mice, SCID ,Lymphocyte Activation ,Lymphoma, T-Cell ,Immunotherapy, Adoptive ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Antigen ,Mice, Inbred NOD ,medicine ,Tumor Cells, Cultured ,Animals ,Humans ,Receptor ,Autocrine signalling ,Antigen-presenting cell ,Cell Proliferation ,Mice, Knockout ,Immunity, Cellular ,Mice, Inbred BALB C ,Receptors, Chimeric Antigen ,Cell growth ,business.industry ,Hematology ,Immunotherapy ,Xenograft Model Antitumor Assays ,Chimeric antigen receptor ,Mice, Inbred C57BL ,030104 developmental biology ,Cytokine ,Oncology ,030220 oncology & carcinogenesis ,Myeloid Differentiation Factor 88 ,Cancer research ,Female ,business ,human activities ,Interleukin-1 - Abstract
Chimeric antigen receptor (CAR) T-cell therapy has shown remarkable responses in B-cell malignancies. However, many patients suffer from limited response and tumor relapse due to lack of persisting CAR T cells and immune escape. These clinical challenges have compromised the long-term efficacy of CAR T-cell therapy and call for the development of novel CAR designs. We demonstrated that CAR T cells secreting a cytokine interleukin-36γ (IL-36γ) showed significantly improved CAR T-cell expansion and persistence, and resulted in superior tumor eradication compared with conventional CAR T cells. The enhanced cellular function by IL-36γ was mediated through an autocrine manner. In addition, activation of endogenous antigen-presenting cells (APCs) and T cells by IL-36γ aided the formation of a secondary antitumor response, which delayed the progression of antigen-negative tumor challenge. Together, our data provide preclinical evidence to support the translation of this design for an improved CAR T-cell-mediated antitumor response.
- Published
- 2020
34. Early experience using salvage radiotherapy for relapsed/refractory non‐Hodgkin lymphomas after CD19 chimeric antigen receptor (CAR) T cell therapy
- Author
-
Sergio Giralt, Michael Scordo, Craig S. Sauter, Brandon S. Imber, Jae H. Park, Connie Lee Batlevi, Gunjan L. Shah, M. Lia Palomba, Miguel-Angel Perales, Michel Sadelain, Carl J. DeSelm, Parastoo B. Dahi, Joachim Yahalom, and Renier J. Brentjens
- Subjects
Adult ,Male ,Oncology ,medicine.medical_specialty ,Allogeneic transplantation ,Subsequent Relapse ,medicine.medical_treatment ,T cell ,Antigens, CD19 ,Cell- and Tissue-Based Therapy ,Salvage therapy ,Immunotherapy, Adoptive ,Article ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,Aged ,Salvage Therapy ,Receptors, Chimeric Antigen ,business.industry ,Lymphoma, Non-Hodgkin ,Hematology ,Middle Aged ,medicine.disease ,Chimeric antigen receptor ,Radiation therapy ,Transplantation ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Female ,Neoplasm Recurrence, Local ,business ,human activities ,Diffuse large B-cell lymphoma ,030215 immunology - Abstract
Radiotherapy is potentially an important salvage strategy post-chimeric antigen receptor T cell therapy (CART), but limited data exist. We reviewed 14 patients treated with salvage radiation post-CART progression (SRT). Most received SRT for first post-CART relapse (71%) to sites previously PET-avid pre-CART (79%). Median overall survival (OS) post-SRT was 10 months. Post-SRT, six localized relapses achieved 100% response (3 = complete, 3 = partial), with improved freedom from subsequent relapse (P = 0·001) and OS (P = 0·004) compared to advanced stage relapses. Three were bridged to allogeneic transplantation; at analysis, all were alive/NED. SRT has diverse utility and can integrate with novel agents or transplantation to attempt durable remissions.
- Published
- 2020
- Full Text
- View/download PDF
35. Preparing for CAR T cell therapy: patient selection, bridging therapies and lymphodepletion
- Author
-
Leila, Amini, Sara K, Silbert, Shannon L, Maude, Loretta J, Nastoupil, Carlos A, Ramos, Renier J, Brentjens, Craig S, Sauter, Nirali N, Shah, and Mohamed, Abou-El-Enein
- Subjects
Patient Selection ,T-Lymphocytes ,Antigens, CD19 ,Humans ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Immunotherapy, Adoptive - Abstract
Chimeric antigen receptor (CAR) T cells have emerged as a potent therapeutic approach for patients with certain haematological cancers, with multiple CAR T cell products currently approved by the FDA for those with relapsed and/or refractory B cell malignancies. However, in order to derive the desired level of effectiveness, patients need to successfully receive the CAR T cell infusion in a timely fashion. This process entails apheresis of the patient's T cells, followed by CAR T cell manufacture. While awaiting infusion at an authorized treatment centre, patients may receive interim disease-directed therapy. Most patients will also receive a course of pre-CAR T cell lymphodepletion, which has emerged as an important factor in enabling durable responses. The time between apheresis and CAR T cell infusion is often not a simple journey, with each milestone being a critical step that can have important downstream consequences for the ability to receive the infusion and the strength of clinical responses. In this Review, we provide a summary of the many considerations for preparing patients with B cell non-Hodgkin lymphoma or acute lymphoblastic leukaemia for CAR T cell therapy, and outline current limitations and areas for future research.
- Published
- 2022
36. CD19-directed chimeric antigen receptor T cell therapy in Waldenström macroglobulinemia: a preclinical model and initial clinical experience
- Author
-
M Lia Palomba, David Qualls, Sebastien Monette, Shenon Sethi, Ahmet Dogan, Mikhail Roshal, Brigitte Senechal, Xiuyan Wang, Isabelle Rivière, Michel Sadelain, Renier J Brentjens, Jae H Park, and Eric L Smith
- Subjects
Pharmacology ,Male ,Cancer Research ,Receptors, Chimeric Antigen ,Immunology ,Antigens, CD19 ,Cell- and Tissue-Based Therapy ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Middle Aged ,Translational Research, Biomedical ,Mice ,Oncology ,Molecular Medicine ,Immunology and Allergy ,Animals ,Humans ,Female ,Waldenstrom Macroglobulinemia ,RC254-282 ,Aged - Abstract
BackgroundWaldenström macroglobulinemia (WM) is an incurable disease and, while treatable, can develop resistance to available therapies and be fatal. Chimeric antigen receptor (CAR) T cell therapy directed against the CD19 antigen has demonstrated efficacy in relapsed or refractory B lymphoid malignancies, and is now approved for B cell acute lymphoblastic leukemia and certain B cell lymphomas. However, CAR T therapy has not been evaluated for use in WM.Methods and resultsWe performed preclinical studies demonstrating CAR T cell activity against WM cells in vitro, and developed an in vivo murine model of WM which demonstrated prolonged survival with use of CAR T therapy. We then report the first three patients with multiply relapsed and refractory WM treated for their disease with CD19-directed CAR T cells on clinical trials. Treatment was well tolerated, and observed toxicities were consistent with those seen in CAR T treatment for other diseases, and no grade 3 or higher cytokine release syndrome or neurotoxicity events occurred. All three patients attained at least a clinical response to treatment, including one minimal residual disease-negative complete response, though all three eventually developed recurrent disease between 3 and 26 months after initial treatment.ConclusionsThis report summarizes preclinical and clinical activity of CD19-directed CAR T therapy in WM, demonstrating early tolerability and efficacy in patients with WM, and representing a possible treatment option in patients with heavily pretreated and relapsed or refractory WM. Larger studies evaluating CAR T therapy in WM are warranted, along with further evaluation into mechanisms of resistance to CAR T therapy.
- Published
- 2022
37. Engineering CAR-T Cells to Activate Small-Molecule Drugs in situ
- Author
-
Thomas J. Gardner, J. Peter Lee, Christopher M. Bourne, Dinali Wijewarnasuriya, Nihar Kinarivala, Keifer G. Kurtz, Broderick C. Corless, Megan M. Dacek, Aaron Y. Chang, George Mo, Kha M. Nguyen, Renier J. Brentjens, Derek S. Tan, and David A. Scheinberg
- Subjects
Male ,Receptors, Chimeric Antigen ,T-Lymphocytes ,Antineoplastic Agents ,Neoplasms, Experimental ,Cell Biology ,Xenograft Model Antitumor Assays ,Article ,Mice, Inbred C57BL ,Mice ,Drug Delivery Systems ,HEK293 Cells ,Neoplasms ,Tumor Microenvironment ,Animals ,Humans ,Female ,Prodrugs ,Molecular Biology - Abstract
Chimeric Antigen Receptor (CAR)-T cells represent a major breakthrough in cancer therapy, wherein a patient’s own T cells are engineered to recognize a tumor antigen, resulting in activation of a local cytotoxic immune response. However, CAR-T cell therapies are currently limited to the treatment of B-cell cancers and their effectiveness is hindered by resistance from antigen-negative tumor cells, immunosuppression in the tumor microenvironment, eventual exhaustion of T-cell immunologic functions, and frequent severe toxicities. To overcome these problems, we have developed a novel class of CAR-T cells engineered to express an enzyme that activates a systemically-administered small-molecule prodrug in situ at a tumor site. We show that these Synthetic Enzyme-Armed KillER (SEAKER) cells exhibit enhanced anticancer activity with small-molecule prodrugs, both in vitro and in vivo in mouse tumor models. This modular platform enables combined targeting of cellular and small-molecule therapies to treat cancers and potentially a variety of other diseases.
- Published
- 2021
38. Human cytomegalovirus expands a CD8
- Author
-
Rosa, Sottile, M Kazim, Panjwani, Colleen M, Lau, Anthony F, Daniyan, Kento, Tanaka, Juliet N, Barker, Renier J, Brentjens, Joseph C, Sun, Jean-Benoît, Le Luduec, and Katharine C, Hsu
- Subjects
Killer Cells, Natural ,Repressor Proteins ,Tumor Suppressor Proteins ,Tumor Cells, Cultured ,Cytomegalovirus ,Humans ,CD8-Positive T-Lymphocytes ,Article - Abstract
CD8(+) T cells are critical mediators of adaptive immunity but may also exhibit innate-like properties such as surface expression of NKG2C, an activating receptor typically associated with natural killer (NK) cells. We demonstrate that, similar to NK cells, NKG2C(+)TCRαβ(+)CD8(+) T cells are associated with prior human cytomegalovirus (HCMV) exposure. In addition to expressing several NK cell markers such as CD56 and KIR, NKG2C(+)CD8(+) T cells are oligoclonal and do not upregulate PD-1 even in response to persistent activation. Furthermore, we found that NKG2C(+)CD8(+) T cells from some individuals exhibited strong effector function against leukemia cells and HCMV-infected fibroblasts, which was dictated by both NKG2C and TCR specificity. Transcriptomic analysis revealed that the transcription factor BCL11B, a regulator of T cell developmental fate, is significantly downregulated in NKG2C(+)CD8(+) T cells when compared to conventional NKG2C(−)CD8(+) T cells. BCL11B deletion in conventional CD8(+) T cells resulted in the emergence of a similar innate-like CD56(+)CD94(+)DAP12(+)NKG2C(+)CD45RA(+)CCR7(−)PD-1(−/low) T cell population with activity against HLA-E(+) targets. Based on their intrinsic capacity to recognize diseased cells coupled with lack of PD-1 induction, NKG2C(+)CD8(+) T cells represent a lymphocyte population that resides at the boundary between innate and adaptive immunity, presenting an attractive alternative for cellular therapy, including CAR T-based therapies.
- Published
- 2021
39. Human cytomegalovirus expands a CD8+T cell population with loss ofBCL11Bexpression and gain of NK cell identity
- Author
-
Renier J. Brentjens, Juliet N. Barker, Anthony F. Daniyan, Kento Tanaka, Rosa Sottile, Jean-Benoît Le Luduec, Joseph C. Sun, Colleen M. Lau, Katharine C. Hsu, and M. Kazim Panjwani
- Subjects
Human cytomegalovirus ,education.field_of_study ,BCL11B ,Immunology ,Population ,General Medicine ,Biology ,Acquired immune system ,medicine.disease ,Cell identity ,Cell biology ,Expression (architecture) ,medicine ,Cytotoxic T cell ,education ,CD8 - Abstract
CD8+ T cells not only are critical mediators of adaptive immunity but also may exhibit innate-like properties such as surface expression of NKG2C, an activating receptor typically associated with n...
- Published
- 2021
- Full Text
- View/download PDF
40. CAR T Cells for Mantle Cell Lymphoma: Is it Time to Reshuffle the Deck?
- Author
-
Renier J. Brentjens and Craig S. Sauter
- Subjects
0301 basic medicine ,Cancer Research ,medicine.drug_class ,business.industry ,medicine.disease ,Tyrosine-kinase inhibitor ,03 medical and health sciences ,Safety profile ,030104 developmental biology ,0302 clinical medicine ,Overall response rate ,New england ,Oncology ,030220 oncology & carcinogenesis ,Refractory Mantle Cell Lymphoma ,medicine ,Cancer research ,Mantle cell lymphoma ,Car t cells ,business - Abstract
In the New England Journal of Medicine, Wang et al. report favorable ZUMA-2 trial results of CAR T cells for patients with relapsed and refractory mantle cell lymphoma following prior failed Bruton's tyrosine kinase inhibitor therapy, with an overall response rate of 93% and an expected safety profile.
- Published
- 2020
- Full Text
- View/download PDF
41. Toxicity and response after CD19-specific CAR T-cell therapy in pediatric/young adult relapsed/refractory B-ALL
- Author
-
Christopher J. Forlenza, Brigitte Senechal, Nancy A. Kernan, Susan E. Prockop, Yasmin Khakoo, David A. Williams, Isabelle Riviere, Peter G. Steinherz, Craig S. Sauter, Jaap Jan Boelens, Michel Sadelain, Lewis B. Silverman, Farid Boulad, Glenn Heller, Richard J. O'Reilly, Victoria Szenes, Jae H. Park, Barbara Spitzer, Neerav Shukla, Andrew L. Kung, Rachel Kobos, Steven P. Margossian, Renier J. Brentjens, Xiuyan Wang, Maria Cancio, and Kevin J Curran
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Cyclophosphamide ,medicine.medical_treatment ,Immunology ,Salvage therapy ,Biochemistry ,Gastroenterology ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Chemotherapy ,business.industry ,Cell Biology ,Hematology ,medicine.disease ,Chemotherapy regimen ,Minimal residual disease ,Cytokine release syndrome ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Chimeric Antigen Receptor T-Cell Therapy ,Bone marrow ,business ,medicine.drug - Abstract
Chimeric antigen receptor (CAR) T cells have demonstrated clinical benefit in patients with relapsed/refractory (R/R) B-cell acute lymphoblastic leukemia (B-ALL). We undertook a multicenter clinical trial to determine toxicity, feasibility, and response for this therapy. A total of 25 pediatric/young adult patients (age, 1-22.5 years) with R/R B-ALL were treated with 19-28z CAR T cells. Conditioning chemotherapy included high-dose (3 g/m2) cyclophosphamide (HD-Cy) for 17 patients and low-dose (≤1.5 g/m2) cyclophosphamide (LD-Cy) for 8 patients. Fifteen patients had pretreatment minimal residual disease (MRD
- Published
- 2019
- Full Text
- View/download PDF
42. Engineering strategies to overcome the current roadblocks in CAR T cell therapy
- Author
-
Sarwish Rafiq, Christopher S. Hackett, and Renier J. Brentjens
- Subjects
0301 basic medicine ,Lymphoma ,T-Lymphocytes ,medicine.medical_treatment ,Genetic enhancement ,T cell ,Cell- and Tissue-Based Therapy ,Receptors, Antigen, T-Cell ,Cancer immunotherapy ,Drug development ,Translational research ,Review Article ,Bioinformatics ,03 medical and health sciences ,Gene therapy ,0302 clinical medicine ,Antigen ,Neoplasms ,Humans ,Medicine ,Cell Engineering ,B cell ,Receptors, Chimeric Antigen ,business.industry ,Chimeric antigen receptor ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,business - Abstract
T cells genetically engineered to express chimeric antigen receptors (CARs) have proven — and impressive — therapeutic activity in patients with certain subtypes of B cell leukaemia or lymphoma, with promising efficacy also demonstrated in patients with multiple myeloma. Nevertheless, various barriers restrict the efficacy and/or prevent the widespread use of CAR T cell therapies in these patients as well as in those with other cancers, particularly solid tumours. Key challenges relating to CAR T cells include severe toxicities, restricted trafficking to, infiltration into and activation within tumours, suboptimal persistence in vivo, antigen escape and heterogeneity, and manufacturing issues. The evolution of CAR designs beyond the conventional structures will be necessary to address these limitations and to expand the use of CAR T cells to a wider range of malignancies. Investigators are addressing the current obstacles with a wide range of engineering strategies in order to improve the safety, efficacy and applicability of this therapeutic modality. In this Review, we discuss the innovative designs of novel CAR T cell products that are being developed to increase and expand the clinical benefits of these treatments in patients with diverse cancers., Chimeric antigen receptor (CAR) T cell therapy, the first approved therapeutic approach with a genetic engineering component, holds substantial promise in the treatment of a range of cancers but is nevertheless limited by various challenges, including toxicities, intrinsic and acquired resistance mechanisms, and manufacturing issues. In this Review, the authors describe the innovative approaches to the engineering of CAR T cell products that are providing solutions to these challenges and therefore have the potential to considerably improve the safety and effectiveness of treatment., Key points Chimeric antigen receptor (CAR) T cells have induced remarkable responses in patients with certain haematological malignancies, yet various barriers restrict the efficacy and/or prevent the widespread use of this treatment.Investigators are addressing these challenges with engineering strategies designed to improve the safety, efficacy and applicability of CAR T cell therapy.CARs have modular components, and therefore the optimal molecular design of the CAR can be achieved through many variations of the constituent protein domains.Toxicities currently associated with CAR T cell therapy can be mitigated using engineering strategies to make CAR T cells safer and that potentially broaden the range of tumour-associated antigens that can be targeted by overcoming on-target, off-tumour toxicities.CAR T cell efficacy can be enhanced by using engineering strategies to address the various challenges relating to the unique biology of diverse haematological and solid malignancies.Strategies to address the manufacturing challenges can lead to an improved CAR T cell product for all patients.
- Published
- 2019
- Full Text
- View/download PDF
43. CAR T‐cell therapy: Full speed ahead
- Author
-
David Sermer and Renier J. Brentjens
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,T-Lymphocytes ,medicine.medical_treatment ,Lymphoblastic Leukemia ,Antigens, CD19 ,Receptors, Antigen, T-Cell ,Immunotherapy, Adoptive ,03 medical and health sciences ,0302 clinical medicine ,Antigens, Neoplasm ,Neoplasms ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Animals ,Humans ,Clinical Trials as Topic ,Receptors, Chimeric Antigen ,business.industry ,Research ,Hematology ,General Medicine ,Immunotherapy ,medicine.disease ,Chimeric antigen receptor ,Lymphoma ,Treatment Outcome ,Neoplasms diagnosis ,030220 oncology & carcinogenesis ,New disease ,CAR T-cell therapy ,Car t cells ,Genetic Engineering ,business ,human activities ,030215 immunology - Abstract
Chimeric antigen receptor (CAR) T-cell therapy has dramatically shifted the landscape of treatment for lymphoid malignancies, especially diffuse large B-cell lymphoma (DLBCL) and acute lymphoblastic leukemia (ALL). However, there continue to be significant limitations of this therapy, such as incomplete or nonsustained responses and severe toxicities in a subset of patients. Furthermore, expanding the role of CAR T-cell therapy to new disease types is an important next step. In this review, we will highlight landmark trials for anti-CD19 CAR T cells and first-in-human trials of novel CARs, as well as discuss promising innovative CAR designs that are still undergoing preclinical development. Lastly, we will discuss toxicity and mechanisms of CAR T-cell resistance and failure, as well as potential future treatment approaches to these common issues.
- Published
- 2019
- Full Text
- View/download PDF
44. Modeling anti-CD19 CAR T cell therapy in humanized mice with human immunity and autologous leukemiaResearch in context
- Author
-
Chun-Hui Jin, Jinxing Xia, Sarwish Rafiq, Xin Huang, Zheng Hu, Xianzheng Zhou, Renier J. Brentjens, and Yong-Guang Yang
- Subjects
lcsh:R5-920 ,lcsh:R ,lcsh:Medicine ,lcsh:Medicine (General) - Abstract
Background: Adoptive immunotherapy using T cells expressing chimeric antigen receptors (CARs) targeting CD19 has produced remarkable clinical outcomes. However, much of the mechanisms of action, such as the development of memory responses and sources of immune cytokines, remain elusive largely due to the challenge of characterizing human CAR T cell function in vivo. The lack of a suitable in vivo model also hinders the development of new CAR T cell therapies. Methods: We established a humanized mouse (hu-mouse) model with a functional human immune system and genetically-matched (autologous) primary acute B-lymphoblastic leukemia (B-ALL) that permits modeling of CD19-targeted CAR T cell therapy in immunocompetent hosts without allogeneic or xenogeneic immune responses. Findings: Anti-CD19 CAR T cells were detected in blood of leukemic hu-mice with kinetics and levels similar to those seen in patients receiving CAR T cell therapy. The levels of CAR T cells were correlated inversely with the burden of leukemia cells and positively with the survival times in anti-CD19 CAR T cell-treated leukemic hu-mice. Infusion of anti-CD19 CAR T cells also resulted in rapid production of T cell- and monocyte/macrophage-derived cytokines and an increase in frequency of regulatory T cells as reported in clinical studies. Interpretation: These results provide a proof-of-principle that this novel preclinical model has the potential to be used to model human CAR T cell therapy and facilitate the design of new CARs with improved antitumor activity.
- Published
- 2019
45. Bispecific T-Cell Engaging Antibodies Against MUC16 Demonstrate Efficacy Against Ovarian Cancer in Monotherapy and in Combination With PD-1 and VEGF Inhibition
- Author
-
Oladapo O. Yeku, Thapi Dharma Rao, Ian Laster, Artem Kononenko, Terence J. Purdon, Pei Wang, Ziyou Cui, Hong Liu, Renier J. Brentjens, and David Spriggs
- Subjects
lcsh:Immunologic diseases. Allergy ,Vascular Endothelial Growth Factor A ,Antineoplastic Agents, Hormonal ,Angiogenesis ,MUC16 ,T-Lymphocytes ,medicine.medical_treatment ,T cell ,Programmed Cell Death 1 Receptor ,Immunology ,angiogenesis ,Mice ,Antigen ,Cell Line, Tumor ,Antibodies, Bispecific ,Animals ,Humans ,Immunology and Allergy ,Medicine ,Immune Checkpoint Inhibitors ,Original Research ,Ovarian Neoplasms ,biology ,MUC16ecto ,business.industry ,Antibody-Dependent Cell Cytotoxicity ,Membrane Proteins ,Drug Synergism ,Immunotherapy ,immune checkpoint blockade ,medicine.disease ,VEGF ,Xenograft Model Antitumor Assays ,Immune checkpoint ,Disease Models, Animal ,ovarian cancer ,medicine.anatomical_structure ,Tumor progression ,CA-125 Antigen ,biology.protein ,Cancer research ,Female ,bispecific engagers ,bispecific antibodies ,Antibody ,lcsh:RC581-607 ,business ,Ovarian cancer - Abstract
Immunotherapy for ovarian cancer is an area of intense investigation since the majority of women with relapsed disease develop resistance to conventional cytotoxic therapy. The paucity of safe and validated target antigens has limited the development of clinically relevant antibody-based immunotherapeutics for this disease. Although MUC16 expression is almost universal in High Grade Serous Ovarian Cancers, engagement of the shed circulating MUC16 antigen (CA-125) presents a theoretical risk of systemic activation and toxicity. We designed and evaluated a series of bispecific tandem single-chain variable fragments specific to the retained portion of human MUC16 ectodomain (MUC16ecto) and human CD3. These MUC16ecto- BiTEDs retain binding in the presence of soluble MUC16 (CA-125) and show cytotoxicity against a panel of ovarian cancer cells in vitro. MUC16ecto- BiTEDs delay tumor progression in vivo and significantly prolong survival in a xenograft model of ovarian peritoneal carcinomatosis. This effect was significantly enhanced by antiangiogenic (anti-VEGF) therapy and immune checkpoint inhibition (anti-PD1). However, the combination of BiTEDs with anti-VEGF was superior to combination with anti-PD1, based on findings of decreased peritoneal tumor burden and ascites with the former. This study shows the feasibility and efficacy of MUC16ecto- specific BiTEDs and provides a basis for the combination with anti-VEGF therapy for ovarian cancer.
- Published
- 2021
- Full Text
- View/download PDF
46. Gut microbiome correlates of response and toxicity following anti-CD19 CAR T cell therapy
- Author
-
Melody Smith, Anqi Dai, Guido Ghilardi, Kimberly V. Amelsberg, Sean M. Devlin, Raymone Pajarillo, John B. Slingerland, Silvia Beghi, Pamela S. Herrera, Paul Giardina, Annelie Clurman, Emmanuel Dwomoh, Gabriel Armijo, Antonio L. C. Gomes, Eric R. Littmann, Jonas Schluter, Emily Fontana, Ying Taur, Jae H. Park, Maria Lia Palomba, Elizabeth Halton, Josel Ruiz, Tania Jain, Martina Pennisi, Aishat Olaide Afuye, Miguel-Angel Perales, Craig W. Freyer, Alfred Garfall, Shannon Gier, Sunita Nasta, Daniel Landsburg, James Gerson, Jakub Svoboda, Justin Cross, Elise A. Chong, Sergio Giralt, Saar I. Gill, Isabelle Riviere, David L. Porter, Stephen J. Schuster, Michel Sadelain, Noelle Frey, Renier J. Brentjens, Carl H. June, Eric G. Pamer, Jonathan U. Peled, Andrea Facciabene, Marcel R. M. van den Brink, and Marco Ruella
- Subjects
Receptors, Chimeric Antigen ,Antigens, CD19 ,Humans ,Neurotoxicity Syndromes ,General Medicine ,Prospective Studies ,Immunotherapy, Adoptive ,Article ,General Biochemistry, Genetics and Molecular Biology ,Gastrointestinal Microbiome ,Retrospective Studies - Abstract
Anti-CD19 chimeric antigen receptor (CAR) T cell therapy has led to unprecedented responses in patients with high-risk hematologic malignancies. However, up to 60% of patients still experience disease relapse and up to 80% of patients experience CAR-mediated toxicities, such as cytokine release syndrome or immune effector cell-associated neurotoxicity syndrome. We investigated the role of the intestinal microbiome on these outcomes in a multicenter study of patients with B cell lymphoma and leukemia. We found in a retrospective cohort (n = 228) that exposure to antibiotics, in particular piperacillin/tazobactam, meropenem and imipenem/cilastatin (P-I-M), in the 4 weeks before therapy was associated with worse survival and increased neurotoxicity. In stool samples from a prospective cohort of CAR T cell recipients (n = 48), the fecal microbiome was altered at baseline compared to healthy controls. Stool sample profiling by 16S ribosomal RNA and metagenomic shotgun sequencing revealed that clinical outcomes were associated with differences in specific bacterial taxa and metabolic pathways. Through both untargeted and hypothesis-driven analysis of 16S sequencing data, we identified species within the class Clostridia that were associated with day 100 complete response. We concluded that changes in the intestinal microbiome are associated with clinical outcomes after anti-CD19 CAR T cell therapy in patients with B cell malignancies.
- Published
- 2021
47. Impact of Bridging Chemotherapy on Clinical Outcome of CD19 CAR T Therapy in Adult Acute Lymphoblastic Leukemia
- Author
-
Kevin J. Curran, Brigitte Senechal, Jae H. Park, Yvette Bernal, Xiuyan Wang, Elizabeth Halton, John Pineda, Mithat Gonen, Karlo Perica, Michel Sadelain, Isabelle Rivere, Claudia Diamonte, Renier J. Brentjens, and Jessica Flynn
- Subjects
Oncology ,Adult ,Cancer Research ,medicine.medical_specialty ,Bridging (networking) ,medicine.medical_treatment ,Antigens, CD19 ,MEDLINE ,Outcome (game theory) ,Immunotherapy, Adoptive ,CD19 ,Article ,Text mining ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Chemotherapy ,biology ,business.industry ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Prognosis ,Combined Modality Therapy ,Survival Rate ,biology.protein ,Adult Acute Lymphoblastic Leukemia ,Car t cells ,business - Published
- 2021
48. Depletion of high-content CD14
- Author
-
Xiuyan, Wang, Oriana, Borquez-Ojeda, Jolanta, Stefanski, Fang, Du, Jinrong, Qu, Jagrutiben, Chaudhari, Keyur, Thummar, Mingzhu, Zhu, Ling-Bo, Shen, Melanie, Hall, Paridhi, Gautam, Yongzeng, Wang, Brigitte, Sénéchal, Devanjan, Sikder, Prasad S, Adusumilli, Renier J, Brentjens, Kevin, Curran, Mark B, Geyer, Sham, Mailankhody, Roisin, O'Cearbhaill, Jae H, Park, Craig, Sauter, Susan, Slovin, Eric L, Smith, and Isabelle, Rivière
- Subjects
cGMP ,CAR T cell ,clinical grade ,large-scale manufacturing ,monocyte depletion ,Original Article - Abstract
With the US Food and Drug Administration (FDA) approval of four CD19- and one BCMA-targeted chimeric antigen receptor (CAR) therapy for B cell malignancies, CAR T cell therapy has finally reached the status of a medicinal product. The successful manufacturing of autologous CAR T cell products is a key requirement for this promising treatment modality. By analyzing the composition of 214 apheresis products from 210 subjects across eight disease indications, we found that high CD14+ cell content poses a challenge for manufacturing CAR T cells, especially in patients with non-Hodgkin’s lymphoma and multiple myeloma caused by the non-specific phagocytosis of the magnetic beads used to activate CD3+ T cells. We demonstrated that monocyte depletion via rapid plastic surface adhesion significantly reduces the CD14+ monocyte content in the apheresis products and simultaneously boosts the CD3+ content. We established a 40% CD14+ threshold for the stratification of apheresis products across nine clinical trials and demonstrated the effectiveness of this procedure by comparing manufacturing runs in two phase 1 clinical trials. Our study suggests that CD14+ content should be monitored in apheresis products, and that the manufacturing of CAR T cells should incorporate a step that lessens the CD14+ cell content in apheresis products containing more than 40% to maximize the production success., Graphical abstract, By analyzing 214 apheresis products and CAR T cell manufacturing runs across multiple disease indications, IR and colleagues have established a threshold of 40% CD14+ cell content as the trigger for the introduction of a rapid plastic adherence monocyte depletion step to enable a 97% manufacturing success rate.
- Published
- 2021
49. Tumor derived UBR5 promotes ovarian cancer growth and metastasis through inducing immunosuppressive macrophages
- Author
-
Briana G. Nixon, Xiaojing Ma, Huan Wang, Mei Song, Junhua Mai, Oladapo Yeku, Tuo Zhang, Haifa Shen, Renier J. Brentjens, Sarwish Rafiq, Xue Dong, Terence J. Purdon, Lijing Zhu, Ziqi Yu, and Ming O. Li
- Subjects
0301 basic medicine ,Chemokine ,medicine.medical_treatment ,General Physics and Astronomy ,Carcinoma, Ovarian Epithelial ,Immunotherapy, Adoptive ,Metastasis ,Mice ,0302 clinical medicine ,Tumor Microenvironment ,Immune Checkpoint Inhibitors ,Peritoneal Neoplasms ,Cancer ,Mice, Knockout ,Ovarian Neoplasms ,Multidisciplinary ,Receptors, Chimeric Antigen ,biology ,Ascites ,Tumor-Derived ,Middle Aged ,Prognosis ,Ubiquitin ligase ,Gene Expression Regulation, Neoplastic ,030220 oncology & carcinogenesis ,embryonic structures ,Disease Progression ,Female ,Adult ,Cell biology ,Science ,Ubiquitin-Protein Ligases ,Immunology ,Primary Cell Culture ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Cell Line, Tumor ,Spheroids, Cellular ,Paracrine Communication ,medicine ,Animals ,Humans ,Cell adhesion ,Aged ,Tumor microenvironment ,business.industry ,General Chemistry ,Immunotherapy ,medicine.disease ,Disease Models, Animal ,030104 developmental biology ,biology.protein ,Cancer research ,Macrophages, Peritoneal ,Tumor Escape ,Ovarian cancer ,business - Abstract
Immunosuppressive tumor microenvironment (TME) and ascites-derived spheroids in ovarian cancer (OC) facilitate tumor growth and progression, and also pose major obstacles for cancer therapy. The molecular pathways involved in the OC-TME interactions, how the crosstalk impinges on OC aggression and chemoresistance are not well-characterized. Here, we demonstrate that tumor-derived UBR5, an E3 ligase overexpressed in human OC associated with poor prognosis, is essential for OC progression principally by promoting tumor-associated macrophage recruitment and activation via key chemokines and cytokines. UBR5 is also required to sustain cell-intrinsic β-catenin-mediated signaling to promote cellular adhesion/colonization and organoid formation by controlling the p53 protein level. OC-specific targeting of UBR5 strongly augments the survival benefit of conventional chemotherapy and immunotherapies. This work provides mechanistic insights into the novel oncogene-like functions of UBR5 in regulating the OC-TME crosstalk and suggests that UBR5 is a potential therapeutic target in OC treatment for modulating the TME and cancer stemness., Ovarian cancer cells often metastasize to the peritoneal cavity, forming spheroid-like structures and promoting a highly immunosuppressive tumor microenvironment. Here, the authors show that the ubiquitin ligase UBR5 is required for ovarian cancer growth and metastasis, sustaining spheroid formation and the infiltration of immunosuppressive tumor associated macrophages.
- Published
- 2020
50. CD103+ cDC1 and endogenous CD8+ T cells are necessary for improved CD40L-overexpressing CAR T cell antitumor function
- Author
-
Winson Cai, Andrea V. Lopez, Anthony F. Daniyan, Renier J. Brentjens, Xinghuo Li, and Nicholas F. Kuhn
- Subjects
0301 basic medicine ,Lymphoma ,Adoptive ,General Physics and Astronomy ,Gene Expression ,Adaptive Immunity ,CD8-Positive T-Lymphocytes ,Immunotherapy, Adoptive ,Mice ,0302 clinical medicine ,Receptors ,Cytotoxic T cell ,Innate ,CD8-positive T cells ,Inbred BALB C ,Mice, Knockout ,Antigen Presentation ,Mice, Inbred BALB C ,Multidisciplinary ,CD11b Antigen ,Receptors, Chimeric Antigen ,hemic and immune systems ,Acquired immune system ,CD ,medicine.anatomical_structure ,Basic-Leucine Zipper Transcription Factors ,Conventional dendritic cells ,030220 oncology & carcinogenesis ,Tumour immunology ,Female ,Immunotherapy ,Integrin alpha Chains ,Biotechnology ,Lymphoma, B-Cell ,Science ,T cell ,Knockout ,1.1 Normal biological development and functioning ,Antigen presentation ,CD40 Ligand ,chemical and pharmacologic phenomena ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Immunophenotyping ,Vaccine Related ,03 medical and health sciences ,Immune system ,Antigen ,Underpinning research ,Antigens, CD ,medicine ,Animals ,Antigens ,CD40 ,Inflammatory and immune system ,B-Cell ,Immunity ,Chimeric Antigen ,General Chemistry ,Dendritic Cells ,Immunity, Innate ,Repressor Proteins ,030104 developmental biology ,Cancer research ,biology.protein ,Immunization ,CD8 ,Neoplasm Transplantation - Abstract
While effective in specific settings, adoptive chimeric antigen receptor (CAR) T cell therapy for cancer requires further improvement and optimization. Our previous results show that CD40L-overexpressing CAR T cells mobilize endogenous immune effectors, resulting in improved antitumor immunity. However, the cell populations required for this protective effect remain to be identified. Here we show, by analyzing Batf3−/− mice lacking the CD103+ conventional dendritic cell type 1 (cDC1) subpopulation important for antigen cross-presentation, that CD40L-overexpressing CAR T cells elicit an impaired antitumor response in the absence of cDC1s. We further find that CD40L-overexpressing CAR T cells stimulate tumor-resident CD11b−CD103− double-negative (DN) cDCs to proliferate and differentiate into cDC1s in wild-type mice. Finally, re-challenge experiments show that endogenous CD8+ T cells are required for protective antitumor memory in this setting. Our findings thus demonstrate the stimulatory effect of CD40L-overexpressing CAR T cells on innate and adaptive immune cells, and provide a rationale for using CD40L-overexpressing CAR T cells to improve immunotherapy responses., CD40L-expressing chimeric antigen receptor (CAR) T cells show enhanced anti-tumor immunity, but the cellular mechanisms are still unclear. Here we show, by analyzing mice deficient of conventional dendritic cell type 1 (cDC1) that cDC1s are induced by CD40L+ CAR T cells to prime endogenous CD8 T cells for a stronger anti-tumor immune response.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.