71 results on '"Roddy S. O’Connor"'
Search Results
2. Enhancing Chimeric Antigen Receptor T Cell Anti-tumor Function through Advanced Media Design
- Author
-
Saba Ghassemi, Francisco J. Martinez-Becerra, Alyssa M. Master, Sarah A. Richman, David Heo, John Leferovich, Yitao Tu, Juan Carlos García-Cañaveras, Asma Ayari, Yinan Lu, Ai Wang, Joshua D. Rabinowitz, Michael C. Milone, Carl H. June, and Roddy S. O’Connor
- Subjects
Genetics ,QH426-470 ,Cytology ,QH573-671 - Abstract
Effective chimeric antigen receptor (CAR)-T cell therapy is dependent on optimal cell culture methods conducive to the activation and expansion of T cells ex vivo, as well as infection with CAR. Media formulations used in CAR-T cell manufacturing have not been optimized for gene delivery, cell expansion, and overall potency. Bioactive components and derivatives that support the generation of functionally-competent T cell progeny with long-lasting persistence are largely undefined. Current media formulations rely on fetal bovine serum (FBS) or human serum (HS), which suffer from a lack of consistency or supply issues. We recognize that components of blood cellular fractions that are absent in serum may have therapeutic value. Here we investigate whether a concentrated growth factor extract, purified from human transfusion grade whole blood fractions, and marketed as PhysiologixTM xeno-free (XF) hGFC (Phx), supports CAR-T cell expansion and function. We show that Phx supports T cell proliferation in clinical and research-grade media. We also show that Phx treatment enhances lentiviral-mediated gene expression across a wide range of multiplicity of infections (MOIs). We compared the ability of anti-GD-2 CAR-T cells expanded ex vivo in medium conditioned with either Phx or HS to clear tumor burden in a human xenograft model of neuroblastoma. We show that T cells expanded in Phx have superior engraftment and potency in vivo, as well as CAR-induced cytolytic activity in vitro. Metabolomic profiling revealed several factors unique to Phx that may have relevance for CAR-T cell preclinical discovery, process development, and manufacturing. In particular, we show that carnosine, a biogenic amine modestly enriched in Phx relative to HS, enhances lentiviral gene delivery in activated T cells. By limiting extracellular acidification, carnosine enhances the metabolic fitness of T cells, shifting their metabolic profile from an acidic, stressed state toward an oxidative, energetic state. These findings are very informative regarding potential derivatives to include in medium customized for gene delivery and overall potency for T cell adoptive immunotherapies.
- Published
- 2020
- Full Text
- View/download PDF
3. Recent Metabolic Advances for Preventing and Treating Acute and Chronic Graft Versus Host Disease
- Author
-
Fathima A. Mohamed, Govindarajan Thangavelu, Stephanie Y. Rhee, Peter T. Sage, Roddy S. O’Connor, Jeffrey C. Rathmell, and Bruce R. Blazar
- Subjects
metabolism ,graft-versus-host disease ,intestinal epithelial cells ,alloreactive T-cells ,graft-versus-tumor ,Immunologic diseases. Allergy ,RC581-607 - Abstract
The therapeutic efficacy of allogeneic hematopoietic stem cell transplantation (allo-HSCT) is limited by the development of graft-versus-host disease (GVHD). In GVHD, rigorous pre-conditioning regimen resets the immune landscape and inflammatory milieu causing immune dysregulation, characterized by an expansion of alloreactive cells and a reduction in immune regulatory cells. In acute GVHD (aGVHD), the release of damage- and pathogen- associated molecular patterns from damaged tissue caused by the conditioning regimen sets the stage for T cell priming, activation and expansion further exacerbating tissue injury and organ damage, particularly in the gastrointestinal tract. Studies have shown that donor T cells utilize multiple energetic and biosynthetic pathways to mediate GVHD that can be distinct from the pathways used by regulatory T cells for their suppressive function. In chronic GVHD (cGVHD), donor T cells may differentiate into IL-21 producing T follicular helper cells or tissue resident T helper cells that cooperate with germinal center B cells or memory B cells, respectively, to produce allo- and auto-reactive antibodies with subsequent tissue fibrosis. Alternatively, donor T cells can become IFN- γ/IL-17 cytokine expressing T cells that mediate sclerodermatous skin injury. Patients refractory to the first line standard regimens for GVHD treatment have a poor prognosis indicating an urgent need for new therapies to restore the balance between effector and regulatory immune cells while preserving the beneficial graft-versus-tumor effect. Emerging data points toward a role for metabolism in regulating these allo- and auto-immune responses. Here, we will discuss the preclinical and clinical data available on the distinct metabolic demands of acute and chronic GVHD and recent efforts in identifying therapeutic targets using metabolomics. Another dimension of this review will examine the changing microbiome after allo-HSCT and the role of microbial metabolites such as short chain fatty acids and long chain fatty acids on regulating immune responses. Lastly, we will examine the metabolic implications of coinhibitory pathway blockade and cellular therapies in allo-HSCT. In conclusion, greater understanding of metabolic pathways involved in immune cell dysregulation during allo-HSCT may pave the way to provide novel therapies to prevent and treat GVHD.
- Published
- 2021
- Full Text
- View/download PDF
4. Case Report: Prolonged Survival Following EGFRvIII CAR T Cell Treatment for Recurrent Glioblastoma
- Author
-
Joseph S. Durgin, Fraser Henderson, MacLean P. Nasrallah, Suyash Mohan, Sumei Wang, Simon F. Lacey, Jan Joseph Melenhorst, Arati S. Desai, John Y. K. Lee, Marcela V. Maus, Carl H. June, Steven Brem, Roddy S. O’Connor, Zev Binder, and Donald M. O’Rourke
- Subjects
CAR T cell therapy ,glioblastoma ,EGFRvIII ,recurrent glioblastoma (rGBM) ,CAR (chimeric antigen receptor) ,perfusion imaging ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Autologous chimeric antigen receptor (CAR) T cells targeted to epidermal growth factor receptor variant III (CAR T-EGFRvIII) have been developed and administered experimentally to treat patients with IDH1 wildtype recurrent glioblastoma (rGBM) (NCT02209376). We report the case of a 59-year-old patient who received a single peripheral infusion of CAR T-EGFRvIII cells and survived 36 months after disease recurrence, exceeding expected survival for recurrent glioblastoma. Post-infusion histopathologic analysis of tissue obtained during a second stage surgical resection revealed immunosuppressive adaptive changes in the tumor tissue as well as reduced EGFRvIII expression. Serial brain imaging demonstrated a significant reduction in relative cerebral blood volume (rCBV), a measure strongly associated with tumor proliferative activity, at early time points following CAR T treatment. Notably, CAR T-EGFRvIII cells persisted in her peripheral circulation during 29 months of follow-up, the longest period of CAR T persistence reported in GBM trials to date. These findings in a long-term survivor show that peripherally administered CAR T-EGFRvIII cells can persist for years in the circulation and suggest that this cell therapy approach could be optimized to achieve broader efficacy in recurrent GBM patients.
- Published
- 2021
- Full Text
- View/download PDF
5. Quantification of lactoyl-CoA (lactyl-CoA) by liquid chromatography mass spectrometry in mammalian cells and tissues
- Author
-
Erika L. Varner, Sophie Trefely, David Bartee, Eliana von Krusenstiern, Luke Izzo, Carmen Bekeova, Roddy S. O'Connor, Erin L. Seifert, Kathryn E. Wellen, Jordan L. Meier, and Nathaniel W. Snyder
- Subjects
metabolism ,lactoyl-coa ,lactyl-coa ,high resolution ,lc-hrms ,Biology (General) ,QH301-705.5 - Abstract
Lysine lactoylation is a recently described protein post-translational modification (PTM). However, the biochemical pathways responsible for this acylation remain unclear. Two metabolite-dependent mechanisms have been proposed: enzymatic histone lysine lactoylation derived from lactoyl-coenzyme A (lactoyl-CoA, also termed lactyl-CoA), and non-enzymatic lysine lactoylation resulting from acyl-transfer via lactoyl-glutathione. While the former has precedent in the form of enzyme-catalysed lysine acylation, the lactoyl-CoA metabolite has not been previously quantified in mammalian systems. Here, we use liquid chromatography–high-resolution mass spectrometry (LC-HRMS) together with a synthetic standard to detect and validate the presence of lactoyl-CoA in cell and tissue samples. Conducting a retrospective analysis of data from previously analysed samples revealed the presence of lactoyl-CoA in diverse cell and tissue contexts. In addition, we describe a biosynthetic route to generate 13C315N1-isotopically labelled lactoyl-CoA, providing a co-eluting internal standard for analysis of this metabolite. We estimate lactoyl-CoA concentrations of 1.14 × 10−8 pmol per cell in cell culture and 0.0172 pmol mg−1 tissue wet weight in mouse heart. These levels are similar to crotonyl-CoA, but between 20 and 350 times lower than predominant acyl-CoAs such as acetyl-, propionyl- and succinyl-CoA. Overall our studies provide the first quantitative measurements of lactoyl-CoA in metazoans, and provide a methodological foundation for the interrogation of this novel metabolite in biology and disease.
- Published
- 2020
- Full Text
- View/download PDF
6. CAR T-Cells Depend on the Coupling of NADH Oxidation with ATP Production
- Author
-
Juan C. Garcia-Canaveras, David Heo, Sophie Trefely, John Leferovich, Chong Xu, Benjamin I. Philipson, Saba Ghassemi, Michael C. Milone, Edmund K. Moon, Nathaniel W. Snyder, Carl H. June, Joshua D. Rabinowitz, and Roddy S. O’Connor
- Subjects
armor CAR T-cells ,Lactobacillus brevis NADH oxidase ,LDHA ,Cytology ,QH573-671 - Abstract
The metabolic milieu of solid tumors provides a barrier to chimeric antigen receptor (CAR) T-cell therapies. Excessive lactate or hypoxia suppresses T-cell growth, through mechanisms including NADH buildup and the depletion of oxidized metabolites. NADH is converted into NAD+ by the enzyme Lactobacillus brevis NADH Oxidase (LbNOX), which mimics the oxidative function of the electron transport chain without generating ATP. Here we determine if LbNOX promotes human CAR T-cell metabolic activity and antitumor efficacy. CAR T-cells expressing LbNOX have enhanced oxygen as well as lactate consumption and increased pyruvate production. LbNOX renders CAR T-cells resilient to lactate dehydrogenase inhibition. But in vivo in a model of mesothelioma, CAR T-cell’s expressing LbNOX showed no increased antitumor efficacy over control CAR T-cells. We hypothesize that T cells in hostile environments face dual metabolic stressors of excessive NADH and insufficient ATP production. Accordingly, futile T-cell NADH oxidation by LbNOX is insufficient to promote tumor clearance.
- Published
- 2021
- Full Text
- View/download PDF
7. Beyond the Lactate Paradox: How Lactate and Acidity Impact T Cell Therapies against Cancer
- Author
-
Violet Y. Tu, Asma Ayari, and Roddy S. O’Connor
- Subjects
lactic acid ,lactate ,acidosis ,acidic ,TME ,CAR T-cells ,Immunologic diseases. Allergy ,RC581-607 - Abstract
T cell therapies, including CAR T cells, have proven more effective in hematologic malignancies than solid tumors, where the local metabolic environment is distinctly immunosuppressive. In particular, the acidic and hypoxic features of the tumor microenvironment (TME) present a unique challenge for T cells. Local metabolism is an important consideration for activated T cells as they undergo bursts of migration, proliferation and differentiation in hostile soil. Tumor cells and activated T cells both produce lactic acid at high rates. The role of lactic acid in T cell biology is complex, as lactate is an often-neglected carbon source that can fuel TCA anaplerosis. Circulating lactate is also an important means to regulate redox balance. In hypoxic tumors, lactate is immune-suppressive. Here, we discuss how intrinsic- (T cells) as well as extrinsic (tumor cells and micro-environmental)-derived metabolic factors, including lactate, suppress the ability of antigen-specific T cells to eradicate tumors. Finally, we introduce recent discoveries that target the TME in order to potentiate T cell-based therapies against cancer.
- Published
- 2021
- Full Text
- View/download PDF
8. Current Status and Challenges of Vaccination Therapy for Glioblastoma
- Author
-
Hamed Hosseinalizadeh, Mohammad Rahmati, Ammar Ebrahimi, and Roddy S. O'Connor
- Subjects
Cancer Research ,Oncology - Abstract
Glioblastoma (GBM), also known as grade IV astrocytoma, is the most common and deadly type of central nervous system malignancy in adults. Despite significant breakthroughs in current GBM treatments such as surgery, radiotherapy, and chemotherapy, the prognosis for late-stage glioblastoma remains bleak due to tumor recurrence following surgical resection. The poor prognosis highlights the evident and pressing need for more efficient and targeted treatment. Vaccination has successfully treated patients with advanced colorectal and lung cancer. Therefore, the potential value of using tumor vaccines in treating glioblastoma is increasingly discussed as a monotherapy or in combination with other cellular immunotherapies. Cancer vaccination includes both passive administration of monoclonal antibodies and active vaccination procedures to activate, boost, or bias antitumor immunity against cancer cells. This article focuses on active immunotherapy with peptide, genetic (DNA, mRNA), and cell-based vaccines in treating GBM and reviews the various treatment approaches currently being tested. Although the ease of synthesis, relative safety, and ability to elicit tumor-specific immune responses have made these vaccines an invaluable tool for cancer treatment, more extensive cohort studies and better guidelines are needed to improve the efficacy of these vaccines in anti-GBM therapy.
- Published
- 2023
9. Supplementary Figure 1 from Current Status and Challenges of Vaccination Therapy for Glioblastoma
- Author
-
Roddy S. O'Connor, Ammar Ebrahimi, Mohammad Rahmati, and Hamed Hosseinalizadeh
- Abstract
Current status and challenges of vaccination therapy for glioblastoma.
- Published
- 2023
10. Data from Reducing Ex Vivo Culture Improves the Antileukemic Activity of Chimeric Antigen Receptor (CAR) T Cells
- Author
-
Michael C. Milone, J. Joseph Melenhorst, Carl H. June, Stephan A. Grupp, Bruce L. Levine, Simon F. Lacey, John Leferovich, Changfeng Zhang, Felipe Bedoya, Megan M. Davis, Stefan M. Lundh, David M. Barrett, John Scholler, Prachi R. Patel, Joseph A. Fraietta, Roddy S. O'Connor, Selene Nunez-Cruz, and Saba Ghassemi
- Abstract
The success of chimeric antigen receptor (CAR)–mediated immunotherapy in acute lymphoblastic leukemia (ALL) highlights the potential of T-cell therapies with directed cytotoxicity against specific tumor antigens. The efficacy of CAR T-cell therapy depends on the engraftment and persistence of T cells following adoptive transfer. Most protocols for T-cell engineering routinely expand T cells ex vivo for 9 to 14 days. Because the potential for engraftment and persistence is related to the state of T-cell differentiation, we hypothesized that reducing the duration of ex vivo culture would limit differentiation and enhance the efficacy of CAR T-cell therapy. We demonstrated that T cells with a CAR-targeting CD19 (CART19) exhibited less differentiation and enhanced effector function in vitro when harvested from cultures at earlier (day 3 or 5) compared with later (day 9) timepoints. We then compared the therapeutic potential of early versus late harvested CART19 in a murine xenograft model of ALL and showed that the antileukemic activity inversely correlated with ex vivo culture time: day 3 harvested cells showed robust tumor control despite using a 6-fold lower dose of CART19, whereas day 9 cells failed to control leukemia at limited cell doses. We also demonstrated the feasibility of an abbreviated culture in a large-scale current good manufacturing practice–compliant process. Limiting the interval between T-cell isolation and CAR treatment is critical for patients with rapidly progressing disease. Generating CAR T cells in less time also improves potency, which is central to the effectiveness of these therapies. Cancer Immunol Res; 6(9); 1100–9. ©2018 AACR.
- Published
- 2023
11. Data from Current Status and Challenges of Vaccination Therapy for Glioblastoma
- Author
-
Roddy S. O'Connor, Ammar Ebrahimi, Mohammad Rahmati, and Hamed Hosseinalizadeh
- Abstract
Glioblastoma (GBM), also known as grade IV astrocytoma, is the most common and deadly type of central nervous system malignancy in adults. Despite significant breakthroughs in current GBM treatments such as surgery, radiotherapy, and chemotherapy, the prognosis for late-stage glioblastoma remains bleak due to tumor recurrence following surgical resection. The poor prognosis highlights the evident and pressing need for more efficient and targeted treatment. Vaccination has successfully treated patients with advanced colorectal and lung cancer. Therefore, the potential value of using tumor vaccines in treating glioblastoma is increasingly discussed as a monotherapy or in combination with other cellular immunotherapies. Cancer vaccination includes both passive administration of monoclonal antibodies and active vaccination procedures to activate, boost, or bias antitumor immunity against cancer cells. This article focuses on active immunotherapy with peptide, genetic (DNA, mRNA), and cell-based vaccines in treating GBM and reviews the various treatment approaches currently being tested. Although the ease of synthesis, relative safety, and ability to elicit tumor-specific immune responses have made these vaccines an invaluable tool for cancer treatment, more extensive cohort studies and better guidelines are needed to improve the efficacy of these vaccines in anti-GBM therapy.
- Published
- 2023
12. Supplemental Data from Reducing Ex Vivo Culture Improves the Antileukemic Activity of Chimeric Antigen Receptor (CAR) T Cells
- Author
-
Michael C. Milone, J. Joseph Melenhorst, Carl H. June, Stephan A. Grupp, Bruce L. Levine, Simon F. Lacey, John Leferovich, Changfeng Zhang, Felipe Bedoya, Megan M. Davis, Stefan M. Lundh, David M. Barrett, John Scholler, Prachi R. Patel, Joseph A. Fraietta, Roddy S. O'Connor, Selene Nunez-Cruz, and Saba Ghassemi
- Abstract
Table S1 and Figures S1 -S8
- Published
- 2023
13. Data from Genomic and Clinical Analysis of Amplification of the 13q31 Chromosomal Region in Alveolar Rhabdomyosarcoma: A Report from the Children's Oncology Group
- Author
-
Frederic G. Barr, Julie M. Gastier-Foster, Mandy J. Dunlevy, Chune Zhang, Roddy S. O'Connor, Donna M. Gustafson, Lynette M. Smith, Fenghai Duan, and Jennifer L. Reichek
- Abstract
Purpose: This study determined the molecular characteristics and clinical significance of amplification of the 13q31 chromosomal region in alveolar rhabdomyosarcoma (ARMS), an aggressive pediatric cancer with frequent PAX3-FOXO1 and PAX7-FOXO1 gene fusions.Experimental Design: The 13q31 amplicon was localized in an initial panel of ARMS cases using oligonucleotide arrays. A fluorescence in situ hybridization assay for this localized region was designed, and applied to more ARMS cases to determine the frequency and distribution of amplification. Quantitative reverse transcription-PCR assays were applied to measure gene expression. The clinical significance of copy number and expression was determined with Kaplan–Meier and Cox proportional hazard models.Results: We localized the 13q31 amplicon to a 0.15 Mb region containing the MIR17HG gene encoding the polycistronic microRNA cluster, miR-17–92. This amplicon is present in 23% of ARMS cases with a marked preference for PAX7-FOXO1-positive cases. In tumors with 13q31 amplification, there is significantly increased expression of 5 of 6 microRNA's within the miR-17–92 cluster (miR-17, miR-19a, miR-19b, miR-20a, and miR-92a). In addition, a subset of nonamplified tumors with copy number-independent overexpression of all 6 microRNA's was identified. In clinical analyses, there was a significantly worse outcome associated with increased expression of the 5 microRNA's described above in 13q31-amplified cases when compared to nonamplified cases. There was also an improved outcome in 13q31-amplified cases with lower expression of these microRNA's.Conclusions: 13q31 amplification and expression of the miR-17–92 cluster provide novel markers for identifying good and poor prognostic subsets of PAX7-FOXO1-positive ARMS. Clin Cancer Res; 17(6); 1463–73. ©2011 AACR.
- Published
- 2023
14. Supplementary Data from Genomic and Clinical Analysis of Amplification of the 13q31 Chromosomal Region in Alveolar Rhabdomyosarcoma: A Report from the Children's Oncology Group
- Author
-
Frederic G. Barr, Julie M. Gastier-Foster, Mandy J. Dunlevy, Chune Zhang, Roddy S. O'Connor, Donna M. Gustafson, Lynette M. Smith, Fenghai Duan, and Jennifer L. Reichek
- Abstract
Supplementary Figures S1-S2; Supplementary Tables S1-S2.
- Published
- 2023
15. Supplementary Materials from Itacitinib (INCB039110), a JAK1 Inhibitor, Reduces Cytokines Associated with Cytokine Release Syndrome Induced by CAR T-cell Therapy
- Author
-
Paul A. Smith, Michael C. Milone, Ahmad Naim, Taisheng Huang, Lisa Truong, Yan-ou Yang, Ashish Juvekar, John Leferovich, Selene Nunez-Cruz, Michael T. Peel, Roddy S. O'Connor, and Eduardo Huarte
- Abstract
Huate et al supplemental figures.
- Published
- 2023
16. Data from Itacitinib (INCB039110), a JAK1 Inhibitor, Reduces Cytokines Associated with Cytokine Release Syndrome Induced by CAR T-cell Therapy
- Author
-
Paul A. Smith, Michael C. Milone, Ahmad Naim, Taisheng Huang, Lisa Truong, Yan-ou Yang, Ashish Juvekar, John Leferovich, Selene Nunez-Cruz, Michael T. Peel, Roddy S. O'Connor, and Eduardo Huarte
- Abstract
Purpose:T cells engineered to express a chimeric antigen receptor (CAR) are a promising cancer immunotherapy. Such targeted therapies have shown long-term relapse-free survival in patients with B-cell leukemia and lymphoma. However, cytokine release syndrome (CRS) represents a serious, potentially life-threatening side effect often associated with CAR T-cell therapy. CRS manifests as a rapid (hyper)immune reaction driven by excessive inflammatory cytokine release, including IFNγ and IL6.Experimental Design:Many cytokines implicated in CRS are known to signal through the JAK-STAT pathway. Here we study the effect of blocking JAK pathway signaling on CAR T-cell proliferation, antitumor activity, and cytokine levels in in vitro and in vivo models.Results:We report that itacitinib, a potent, selective JAK1 inhibitor, was able to significantly and dose-dependently reduce levels of multiple cytokines implicated in CRS in several in vitro and in vivo models. Importantly, we also report that at clinically relevant doses that mimic human JAK1 pharmacologic inhibition, itacitinib did not significantly inhibit proliferation or antitumor killing capacity of three different human CAR T-cell constructs (GD2, EGFR, and CD19). Finally, in an in vivo model, antitumor activity of CD19-CAR T cells adoptively transferred into CD19+ tumor-bearing immunodeficient animals was unabated by oral itacitinib treatment.Conclusions:Together, these data suggest that itacitinib has potential as a prophylactic agent for the prevention of CAR T cell–induced CRS, and a phase II clinical trial of itacitinib for prevention of CRS induced by CAR T-cell therapy has been initiated (NCT04071366).
- Published
- 2023
17. Targeting PARP11 to avert immunosuppression and improve CAR T therapy in solid tumors
- Author
-
Hongru Zhang, Pengfei Yu, Vivek S. Tomar, Xiangjie Chen, Matthew J. Atherton, Zhen Lu, Hong-Guang Zhang, Shifeng Li, Angelica Ortiz, Jun Gui, N. Adrian Leu, Fangxue Yan, Andres Blanco, Mirella L. Meyer-Ficca, Ralph G. Meyer, Daniel P. Beiting, Jinyang Li, Selene Nunez-Cruz, Roddy S. O’Connor, Lexus R. Johnson, Andy J. Minn, Subin S. George, Constantinos Koumenis, J. Alan Diehl, Michael C. Milone, Hui Zheng, and Serge Y. Fuchs
- Subjects
Immunosuppression Therapy ,Cancer Research ,Receptors, Chimeric Antigen ,Oncology ,Neoplasms ,Tumor Microenvironment ,Humans ,Poly(ADP-ribose) Polymerases ,Immunotherapy, Adoptive - Abstract
Evasion of antitumor immunity and resistance to therapies in solid tumors are aided by an immunosuppressive tumor microenvironment (TME). We found that TME factors, such as regulatory T cells and adenosine, downregulated type I interferon receptor IFNAR1 on CD8
- Published
- 2022
18. Enhancing CAR T function with the engineered secretion of C. perfringens neuraminidase
- Author
-
Joseph S. Durgin, Zev A. Binder, Vijay Bhoj, Michael C. Milone, Donald M. O'Rourke, Radhika Thokala, Saba Ghassemi, Roddy S. O’Connor, Lexus R. Johnson, John Leferovich, and Edward Z Song
- Subjects
Adoptive cell transfer ,Clostridium perfringens ,T cell ,Antigens, CD19 ,Cell ,Neuraminidase ,Immunotherapy, Adoptive ,Immune system ,Cell Line, Tumor ,Drug Discovery ,Genetics ,medicine ,Humans ,Molecular Biology ,Pharmacology ,Receptors, Chimeric Antigen ,biology ,Effector ,Chemistry ,Xenograft Model Antitumor Assays ,Immune checkpoint ,Cell biology ,Cytolysis ,medicine.anatomical_structure ,biology.protein ,Molecular Medicine - Abstract
Prior to adoptive transfer, CAR T cells are activated, lentivirally infected with CAR transgenes, and expanded over 9 to 11 days. An unintended consequence of this process is the progressive differentiation of CAR T cells over time in culture. Differentiated T cells engraft poorly, which limits their ability to persist and provide sustained tumor control in hematologic as well as solid tumors. Solid tumors include other barriers to CAR T cell therapies, including immune and metabolic checkpoints that suppress effector function and durability. Sialic acids are ubiquitous surface molecules with known immune checkpoint functions. The enzyme C. perfringens neuraminidase (CpNA) removes sialic acid residues from target cells, with good activity at physiologic conditions. In combination with galactose oxidase (GO), NA has been found to stimulate T cell mitogenesis and cytotoxicity in vitro. Here we determine whether CpNA alone and in combination with GO promotes CAR T cell antitumor efficacy. We show that CpNA restrains CAR T cell differentiation during ex vivo culture, giving rise to progeny with enhanced therapeutic potential. CAR T cells expressing CpNA have superior effector function and cytotoxicity in vitro. In a Nalm-6 xenograft model of leukemia, CAR T cells expressing CpNA show enhanced antitumor efficacy. Arming CAR T cells with CpNA also enhanced tumor control in xenograft models of glioblastoma as well as a syngeneic model of melanoma. Given our findings, we hypothesize that charge repulsion via surface glycans is a regulatory parameter influencing differentiation. As T cells engage target cells within tumors and undergo constitutive activation through their CARs, critical thresholds of negative charge may impede cell-cell interactions underlying synapse formation and cytolysis. Removing the dense pool of negative cell-surface charge with CpNA is an effective approach to limit CAR T cell differentiation and enhance overall persistence and efficacy.
- Published
- 2022
19. Rapid manufacturing of non-activated potent CAR T cells
- Author
-
Saba Ghassemi, Joseph S. Durgin, Selene Nunez-Cruz, Jai Patel, John Leferovich, Marilia Pinzone, Feng Shen, Katherine D. Cummins, Gabriela Plesa, Vito Adrian Cantu, Shantan Reddy, Frederic D. Bushman, Saar I. Gill, Una O’Doherty, Roddy S. O’Connor, and Michael C. Milone
- Subjects
Biomedical Engineering ,Medicine (miscellaneous) ,Bioengineering ,Computer Science Applications ,Biotechnology - Published
- 2022
20. In Like a Lamb; Out Like a Lion: Marching CAR T Cells Toward Enhanced Efficacy in B-ALL
- Author
-
Pouya Safarzadeh Kozani, Roddy S. O’Connor, and Pooria Safarzadeh Kozani
- Subjects
Cancer Research ,T-Lymphocytes ,Antigens, CD19 ,Receptors, Antigen, T-Cell ,T-Cell Antigen Receptor Specificity ,Biology ,Immunotherapy, Adoptive ,Article ,Synthetic biology ,Antigen ,Antigens, Neoplasm ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,medicine ,Animals ,Humans ,Receptors, Chimeric Antigen ,Effector ,Immunity ,Disease Management ,Cancer ,medicine.disease ,Chimeric antigen receptor ,Lymphoma ,Treatment Outcome ,Oncology ,Research Design ,Cancer research ,Tumor Escape ,Mantle cell lymphoma ,Genetic Engineering ,Function (biology) - Abstract
Combining synthetic biology with adoptive T-cell transfer has led to promising advances in the treatment of relapsed/refractory B-cell acute lymphoblastic leukemia (R/R B-ALL), diffuse large B-cell lymphoma (DLBCL), and mantle cell lymphoma (MCL). Chimeric antigen receptors (CARs) are synthetic receptors that redirect T-cell specificity against cancer. CARs include “built-in” signaling domains that reprogram T-cell metabolism, enhance effector function, and support long-term persistence. Despite their success in blood-based malignancies, relapse can occur in CD19-redirected CAR T-cell therapies for several reasons, including poor engraftment, impaired in vivo proliferation, and T-cell senescence. Herein, we explain how subtle alterations in CAR design may overcome barriers to effective adoptive immunotherapy. We also discuss how the physiochemical properties of the single-chain variable fragment (scFv) affect differentiation and persistence. Moreover, we describe innovative advances in CAR engineering and provide insight into the development of humanized scFvs whose proposed benefits include increased persistence and improved clinical outcomes. Tumor cells can evade CAR T-cell–mediated detection and elimination due to the emergence or presence of CD19-negative leukemic cell subpopulations. We also discuss the opportunities and challenges in targeting other B-ALL–associated antigens. Identifying alternate targets is fundamentally necessary to restore the success of CAR T-cell therapies in CD19-negative patients with B-ALL.
- Published
- 2021
21. Itacitinib (INCB039110), a JAK1 Inhibitor, Reduces Cytokines Associated with Cytokine Release Syndrome Induced by CAR T-cell Therapy
- Author
-
Paul A. Smith, Yan-ou Yang, Ashish Juvekar, Roddy S. O’Connor, Michael Peel, Selene Nunez-Cruz, Ahmad Naim, Michael C. Milone, Eduardo Huarte, Taisheng Huang, Lisa Truong, and John Leferovich
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,Apoptosis ,Mice, SCID ,Immunotherapy, Adoptive ,Article ,CD19 ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cancer immunotherapy ,Mice, Inbred NOD ,In vivo ,JAK1 Inhibitor ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Cell Proliferation ,Mice, Inbred BALB C ,biology ,business.industry ,Janus Kinase 1 ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Xenograft Model Antitumor Assays ,Chimeric antigen receptor ,Mice, Inbred C57BL ,Cytokine release syndrome ,Leukemia ,030104 developmental biology ,Cytokine ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Azetidines ,Cytokines ,Female ,Isonicotinic Acids ,Cytokine Release Syndrome ,business - Abstract
Purpose: T cells engineered to express a chimeric antigen receptor (CAR) are a promising cancer immunotherapy. Such targeted therapies have shown long-term relapse-free survival in patients with B-cell leukemia and lymphoma. However, cytokine release syndrome (CRS) represents a serious, potentially life-threatening side effect often associated with CAR T-cell therapy. CRS manifests as a rapid (hyper)immune reaction driven by excessive inflammatory cytokine release, including IFNγ and IL6. Experimental Design: Many cytokines implicated in CRS are known to signal through the JAK-STAT pathway. Here we study the effect of blocking JAK pathway signaling on CAR T-cell proliferation, antitumor activity, and cytokine levels in in vitro and in vivo models. Results: We report that itacitinib, a potent, selective JAK1 inhibitor, was able to significantly and dose-dependently reduce levels of multiple cytokines implicated in CRS in several in vitro and in vivo models. Importantly, we also report that at clinically relevant doses that mimic human JAK1 pharmacologic inhibition, itacitinib did not significantly inhibit proliferation or antitumor killing capacity of three different human CAR T-cell constructs (GD2, EGFR, and CD19). Finally, in an in vivo model, antitumor activity of CD19-CAR T cells adoptively transferred into CD19+ tumor-bearing immunodeficient animals was unabated by oral itacitinib treatment. Conclusions: Together, these data suggest that itacitinib has potential as a prophylactic agent for the prevention of CAR T cell–induced CRS, and a phase II clinical trial of itacitinib for prevention of CRS induced by CAR T-cell therapy has been initiated (NCT04071366).
- Published
- 2020
22. Enhancing Chimeric Antigen Receptor T Cell Anti-tumor Function through Advanced Media Design
- Author
-
Michael C. Milone, Francisco J. Martinez-Becerra, Yinan Lu, Alyssa Master, David Heo, Saba Ghassemi, Joshua D. Rabinowitz, Juan Carlos García-Cañaveras, Ai Wang, Yitao Tu, Carl H. June, Asma Ayari, Roddy S. O’Connor, John Leferovich, and Sarah A. Richman
- Subjects
0301 basic medicine ,lcsh:QH426-470 ,Chemistry ,lcsh:Cytology ,T cell ,Cell ,Gene delivery ,Chimeric antigen receptor ,Article ,Cell biology ,Cell therapy ,03 medical and health sciences ,Cytolysis ,lcsh:Genetics ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Cell culture ,030220 oncology & carcinogenesis ,Genetics ,medicine ,Molecular Medicine ,lcsh:QH573-671 ,Molecular Biology ,Fetal bovine serum - Abstract
Effective chimeric antigen receptor (CAR)-T cell therapy is dependent on optimal cell culture methods conducive to the activation and expansion of T cells ex vivo, as well as infection with CAR. Media formulations used in CAR-T cell manufacturing have not been optimized for gene delivery, cell expansion, and overall potency. Bioactive components and derivatives that support the generation of functionally-competent T cell progeny with long-lasting persistence are largely undefined. Current media formulations rely on fetal bovine serum (FBS) or human serum (HS), which suffer from a lack of consistency or supply issues. We recognize that components of blood cellular fractions that are absent in serum may have therapeutic value. Here we investigate whether a concentrated growth factor extract, purified from human transfusion grade whole blood fractions, and marketed as PhysiologixTM xeno-free (XF) hGFC (Phx), supports CAR-T cell expansion and function. We show that Phx supports T cell proliferation in clinical and research-grade media. We also show that Phx treatment enhances lentiviral-mediated gene expression across a wide range of multiplicity of infections (MOIs). We compared the ability of anti-GD-2 CAR-T cells expanded ex vivo in medium conditioned with either Phx or HS to clear tumor burden in a human xenograft model of neuroblastoma. We show that T cells expanded in Phx have superior engraftment and potency in vivo, as well as CAR-induced cytolytic activity in vitro. Metabolomic profiling revealed several factors unique to Phx that may have relevance for CAR-T cell preclinical discovery, process development, and manufacturing. In particular, we show that carnosine, a biogenic amine modestly enriched in Phx relative to HS, enhances lentiviral gene delivery in activated T cells. By limiting extracellular acidification, carnosine enhances the metabolic fitness of T cells, shifting their metabolic profile from an acidic, stressed state toward an oxidative, energetic state. These findings are very informative regarding potential derivatives to include in medium customized for gene delivery and overall potency for T cell adoptive immunotherapies., Graphical Abstract, Media formulations used in CAR-T cell therapies have not been optimized for gene delivery, cell expansion, and overall potency. Ghassemi et al. report that anti-GD2 CAR-T cells expanded in medium conditioned with a concentrated growth factor extract have superior engraftment and potency in a human xenograft model of neuroblastoma.
- Published
- 2020
23. Humanized Chimeric Antigen Receptor (CAR) T cells
- Author
-
Pouya Safarzadeh, Kozani, Pooria Safarzadeh, Kozani, and Roddy S, O'Connor
- Subjects
Article - Published
- 2022
24. Metabolic and epigenetic orchestration of (CAR) T cell fate and function
- Author
-
Behnia Akbari, Zahra Hosseini, Pardis Shahabinejad, Saba Ghassemi, Hamid Reza Mirzaei, and Roddy S. O'Connor
- Subjects
Cancer Research ,Receptors, Chimeric Antigen ,Oncology ,Neoplasms ,T-Lymphocytes ,Tumor Microenvironment ,Humans ,Antigens, Viral, Tumor ,Immunotherapy, Adoptive ,Epigenesis, Genetic - Abstract
Longevity, functionality, and metabolic fitness are key determinants of chimeric antigen receptor (CAR) T cell efficacy. Activated T cells follow an ordered differentiation program which is facilitated by metabolic adaptations. In response to antigen, T cells undergo a highly-regulated shift to glycolysis. Committing to, and engaging in, glycolysis supports T cell expansion and effector function. Inside tumors, heightened tumor cell metabolism and dysregulated perfusion create a competition for nutrients. As local metabolism supports the differentiation of T cells into functionally-competent progeny, nutrient depletion coupled with persisting antigen can trigger T cell exhaustion. Emerging insights into the barriers impeding CAR T cell function in hostile tumor microenvironments (TME) reveal that metabolic intermediates shape the immune response by influencing epigenetic programs and the control of gene expression. In this review, we discuss recent progress connecting cellular metabolism with epigenetic states in CAR T cells. Given that CAR T cell metabolism can be dynamically regulated, we introduce the concepts of "metabolic-based epigenetic altering" and "epigenetic-based metabolism altering" to restore functional competence in CARTs traversing solid TMEs.
- Published
- 2022
25. CAR T-Cells Depend on the Coupling of NADH Oxidation with ATP Production
- Author
-
Sophie Trefely, John Leferovich, Saba Ghassemi, Joshua D. Rabinowitz, Michael C. Milone, David Heo, Chong Xu, Roddy S. O’Connor, Carl H. June, Edmund K. Moon, Juan Carlos García-Cañaveras, Benjamin Philipson, and Nathaniel W. Snyder
- Subjects
Adult ,Male ,Lactobacillus brevis NADH oxidase ,QH301-705.5 ,T-Lymphocytes ,Levilactobacillus brevis ,Receptors, Antigen, T-Cell ,Oxidative phosphorylation ,Mice, SCID ,armor CAR T-cells ,Article ,LDHA ,chemistry.chemical_compound ,Mice ,Adenosine Triphosphate ,In vivo ,Mice, Inbred NOD ,Multienzyme Complexes ,Lactate dehydrogenase ,medicine ,Animals ,Humans ,NADH, NADPH Oxidoreductases ,Biology (General) ,chemistry.chemical_classification ,biology ,Lactobacillus brevis ,General Medicine ,Hypoxia (medical) ,biology.organism_classification ,NAD ,Electron transport chain ,Enzyme ,Biochemistry ,chemistry ,Female ,NAD+ kinase ,medicine.symptom ,Oxidation-Reduction - Abstract
The metabolic milieu of solid tumors provides a barrier to chimeric antigen receptor (CAR) T-cell therapies. Excessive lactate or hypoxia suppresses T-cell growth, through mechanisms including NADH buildup and the depletion of oxidized metabolites. NADH is converted into NAD+ by the enzyme Lactobacillus brevis NADH Oxidase (LbNOX), which mimics the oxidative function of the electron transport chain without generating ATP. Here we determine if LbNOX promotes human CAR T-cell metabolic activity and antitumor efficacy. CAR T-cells expressing LbNOX have enhanced oxygen as well as lactate consumption and increased pyruvate production. LbNOX renders CAR T-cells resilient to lactate dehydrogenase inhibition. But in vivo in a model of mesothelioma, CAR T-cell’s expressing LbNOX showed no increased antitumor efficacy over control CAR T-cells. We hypothesize that T cells in hostile environments face dual metabolic stressors of excessive NADH and insufficient ATP production. Accordingly, futile T-cell NADH oxidation by LbNOX is insufficient to promote tumor clearance.
- Published
- 2021
26. Beyond the Lactate Paradox: How Lactate and Acidity Impact T Cell Therapies against Cancer
- Author
-
Roddy S. O’Connor, Asma Ayari, and Violet Y. Tu
- Subjects
0301 basic medicine ,CAR T-cells ,T cell ,Immunology ,VISTA ,Review ,LDHA ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Drug Discovery ,Carbon source ,medicine ,Immunology and Allergy ,Acidosis ,Tumor microenvironment ,lactate ,acidic ,Chemistry ,lactic acid ,TME ,Cancer ,Metabolism ,RC581-607 ,medicine.disease ,Lactic acid ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer research ,acidosis ,medicine.symptom ,Car t cells ,Immunologic diseases. Allergy - Abstract
T cell therapies, including CAR T cells, have proven more effective in hematologic malignancies than solid tumors, where the local metabolic environment is distinctly immunosuppressive. In particular, the acidic and hypoxic features of the tumor microenvironment (TME) present a unique challenge for T cells. Local metabolism is an important consideration for activated T cells as they undergo bursts of migration, proliferation and differentiation in hostile soil. Tumor cells and activated T cells both produce lactic acid at high rates. The role of lactic acid in T cell biology is complex, as lactate is an often-neglected carbon source that can fuel TCA anaplerosis. Circulating lactate is also an important means to regulate redox balance. In hypoxic tumors, lactate is immune-suppressive. Here, we discuss how intrinsic- (T cells) as well as extrinsic (tumor cells and micro-environmental)-derived metabolic factors, including lactate, suppress the ability of antigen-specific T cells to eradicate tumors. Finally, we introduce recent discoveries that target the TME in order to potentiate T cell-based therapies against cancer.
- Published
- 2021
27. Metabolic rewiring of macrophages by CpG potentiates clearance of cancer cells and overcomes tumor-expressed CD47-mediated ‘don’t eat me signal’
- Author
-
Mingen Liu, Roddy S. O’Connor, Kathleen Graham, Sophie Trefely, Gregory L. Beatty, and Nathaniel W. Snyder
- Subjects
0301 basic medicine ,ATP citrate lyase ,CpG Oligodeoxynucleotide ,Cell Survival ,Immunology ,CD47 Antigen ,Mice, Transgenic ,Article ,03 medical and health sciences ,0302 clinical medicine ,Phagocytosis ,Lipid biosynthesis ,Cell Line, Tumor ,Immunology and Allergy ,Macrophage ,Animals ,Humans ,Receptor ,Cells, Cultured ,Mice, Knockout ,Chemistry ,CD47 ,Macrophages ,Coculture Techniques ,Cell biology ,Mice, Inbred C57BL ,Pancreatic Neoplasms ,030104 developmental biology ,CpG site ,Oligodeoxyribonucleotides ,Toll-Like Receptor 9 ,Cancer cell ,030215 immunology - Abstract
Macrophages enforce antitumor immunity by engulfing and killing tumor cells. Although these functions are determined by a balance of stimulatory and inhibitory signals, the role of macrophage metabolism is unknown. Here, we study the capacity of macrophages to circumvent inhibitory activity mediated by CD47 on cancer cells. We show that stimulation with a CpG oligodeoxynucleotide, a Toll-like receptor 9 agonist, evokes changes in the central carbon metabolism of macrophages that enable antitumor activity, including engulfment of CD47+ cancer cells. CpG activation engenders a metabolic state that requires fatty acid oxidation and shunting of tricarboxylic acid cycle intermediates for de novo lipid biosynthesis. This integration of metabolic inputs is underpinned by carnitine palmitoyltransferase 1A and adenosine tri-phosphate citrate lyase, which, together, impart macrophages with antitumor potential capable of overcoming inhibitory CD47 on cancer cells. Our findings identify central carbon metabolism to be a novel determinant and potential therapeutic target for stimulating antitumor activity by macrophages. Macrophages have important antitumor functions but can be evaded by tumor-expressed CD47-dependent ‘don’t-eat-me’ signals. Beatty and colleagues demonstrate that the Toll-like receptor 9 ligand CpG metabolically reprograms macrophages to overcome don’t-eat-me signals.
- Published
- 2019
28. Rapid manufacturing of non-activated potent CAR T cells
- Author
-
Saba, Ghassemi, Joseph S, Durgin, Selene, Nunez-Cruz, Jai, Patel, John, Leferovich, Marilia, Pinzone, Feng, Shen, Katherine D, Cummins, Gabriela, Plesa, Vito Adrian, Cantu, Shantan, Reddy, Frederic D, Bushman, Saar I, Gill, Una, O'Doherty, Roddy S, O'Connor, and Michael C, Milone
- Subjects
Mice ,Leukemia ,Receptors, Chimeric Antigen ,T-Lymphocytes ,Animals ,Humans ,Immunotherapy, Adoptive - Abstract
Chimaeric antigen receptor (CAR) T cells can generate durable clinical responses in B-cell haematologic malignancies. The manufacturing of these T cells typically involves their activation, followed by viral transduction and expansion ex vivo for at least 6 days. However, the activation and expansion of CAR T cells leads to their progressive differentiation and the associated loss of anti-leukaemic activity. Here we show that functional CAR T cells can be generated within 24 hours from T cells derived from peripheral blood without the need for T-cell activation or ex vivo expansion, and that the efficiency of viral transduction in this process is substantially influenced by the formulation of the medium and the surface area-to-volume ratio of the culture vessel. In mouse xenograft models of human leukaemias, the rapidly generated non-activated CAR T cells exhibited higher anti-leukaemic in vivo activity per cell than the corresponding activated CAR T cells produced using the standard protocol. The rapid manufacturing of CAR T cells may reduce production costs and broaden their applicability.
- Published
- 2021
29. Re: Human Chimeric Antigen Receptor Macrophages for Cancer Immunotherapy
- Author
-
Benjamin A. Garcia, Saar Gill, Dylan M. Marchione, Konrad Gabrusiewicz, Kimberly Veliz, Xueqing Maggie Lu, Nicholas R. Anderson, Olga Shestova, Miriam Y. Kim, Feng Shen, Stephen R. Wallace, Michael Klichinsky, Xinhe Shan, Carl H. June, Roddy S. O’Connor, Kristin Blouch, Maksim Shestov, Marco Ruella, Yumi Yashiro-Ohtani, Miroslaw Kozlowski, Martha Zeeman, Katherine D. Cummins, Maggie Schmierer, Andrew Best, Nicholas E. Petty, and Saad S. Kenderian
- Subjects
Lung Neoplasms ,Cell Survival ,T cell ,Urology ,medicine.medical_treatment ,Antigen presentation ,Biomedical Engineering ,Bioengineering ,Biology ,Applied Microbiology and Biotechnology ,Immunotherapy, Adoptive ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Antigen ,Cancer immunotherapy ,Cell Line, Tumor ,Neoplasms ,medicine ,Macrophage ,Animals ,Humans ,030304 developmental biology ,0303 health sciences ,Tumor microenvironment ,Microscopy, Video ,business.industry ,Macrophages ,Immunotherapy ,Neoplasms, Experimental ,Chimeric antigen receptor ,medicine.anatomical_structure ,Humanized mouse ,Cancer research ,Molecular Medicine ,business ,030217 neurology & neurosurgery ,Biotechnology - Abstract
Chimeric antigen receptor (CAR) T cell therapy has shown promise in hematologic malignancies, but its application to solid tumors has been challenging1–4. Given the unique effector functions of macrophages and their capacity to penetrate tumors5, we genetically engineered human macrophages with CARs to direct their phagocytic activity against tumors. We found that a chimeric adenoviral vector overcame the inherent resistance of primary human macrophages to genetic manipulation and imparted a sustained pro-inflammatory (M1) phenotype. CAR macrophages (CAR-Ms) demonstrated antigen-specific phagocytosis and tumor clearance in vitro. In two solid tumor xenograft mouse models, a single infusion of human CAR-Ms decreased tumor burden and prolonged overall survival. Characterization of CAR-M activity showed that CAR-Ms expressed pro-inflammatory cytokines and chemokines, converted bystander M2 macrophages to M1, upregulated antigen presentation machinery, recruited and presented antigen to T cells and resisted the effects of immunosuppressive cytokines. In humanized mouse models, CAR-Ms were further shown to induce a pro-inflammatory tumor microenvironment and boost anti-tumor T cell activity. Primary macrophages engineered to express chimeric antigen receptors have anti-tumor activity in humanized mice.
- Published
- 2021
30. Citius, Altius, Fortius: Performance in a Bottle for CAR T-Cells
- Author
-
Roddy S. O’Connor and Asma Ayari
- Subjects
business.product_category ,Chemistry ,Computer graphics (images) ,Bottle ,Car t cells ,business ,Article - Published
- 2020
31. Lingering effects of chemotherapy on mature T cells impair proliferation
- Author
-
Rajat K. Das, Stephan A. Grupp, Roddy S. O’Connor, and David M. Barrett
- Subjects
0301 basic medicine ,Cyclophosphamide ,Immunobiology and Immunotherapy ,medicine.medical_treatment ,T-Lymphocytes ,Immunotherapy, Adoptive ,Cell therapy ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,medicine ,Humans ,Doxorubicin ,Child ,Cell Proliferation ,Chemotherapy ,Receptors, Chimeric Antigen ,business.industry ,Hematology ,Immunotherapy ,Pediatric cancer ,Chimeric antigen receptor ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cytarabine ,Cancer research ,business ,medicine.drug - Abstract
Engineered T-cell therapies have demonstrated impressive clinical responses in patients with hematologic malignancies. Despite this efficacy, many patients have a transient persistence of T cells, which can be correlated with transient clinical response. Translational data on T cells from pediatric cancer patients shows a progressive decline in chimeric antigen receptor (CAR) suitability with cumulative chemotherapy regardless of regimen. We investigated the effects of chemotherapy on surviving T cells in vitro, describing residual deficits unique to each agent including mitochondrial damage and metabolic alterations. In the case of cyclophosphamide but not doxorubicin or cytarabine, these effects could be reversed with N-acetylcysteine. Specifically, we observed that surviving T cells could be stimulated, expanded, and transduced with CARs with preserved short-term cytolytic function but at far lower numbers and with residual metabolic deficits. These data have implications for understanding the effects of chemotherapy on mature T cells later collected for adoptive cell therapy, as chemotherapy-exposed T cells may have lingering dysfunction that affects ex vivo adoptive cell therapy manufacturing techniques and, ultimately, clinical efficacy.
- Published
- 2020
32. Quantification of lactoyl-CoA (lactyl-CoA) by liquid chromatography mass spectrometry in mammalian cells and tissues
- Author
-
Eliana von Krusenstiern, Carmen Bekeova, Nathaniel W. Snyder, Erika L. Varner, Luke Izzo, David Bartee, Erin L. Seifert, Sophie Trefely, Kathryn E. Wellen, Roddy S. O’Connor, and Jordan L. Meier
- Subjects
lactoyl-CoA ,LC-HRMS ,Metabolite ,Immunology ,Lysine ,Biology ,Mass spectrometry ,01 natural sciences ,General Biochemistry, Genetics and Molecular Biology ,Mass Spectrometry ,Acylation ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Liquid chromatography–mass spectrometry ,Animals ,Metabolomics ,lcsh:QH301-705.5 ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,Molecular Structure ,010405 organic chemistry ,lactyl-CoA ,high resolution ,General Neuroscience ,Research ,Metabolism ,0104 chemical sciences ,Metabolic pathway ,Enzyme ,Biochemistry ,chemistry ,lcsh:Biology (General) ,Organ Specificity ,Acyl Coenzyme A ,metabolism ,Biomarkers ,Metabolic Networks and Pathways ,Chromatography, Liquid ,Research Article - Abstract
Lysine lactoylation is a recently described protein post-translational modification (PTM). However, the biochemical pathways responsible for this acylation remain unclear. Two metabolite-dependent mechanisms have been proposed: enzymatic histone lysine lactoylation derived from lactoyl-coenzyme A (lactoyl-CoA, also termed lactyl-CoA), and non-enzymatic lysine lactoylation resulting from acyl-transfer via lactoyl-glutathione. While the former has precedent in the form of enzyme-catalysed lysine acylation, the lactoyl-CoA metabolite has not been previously quantified in mammalian systems. Here, we use liquid chromatography–high-resolution mass spectrometry (LC-HRMS) together with a synthetic standard to detect and validate the presence of lactoyl-CoA in cell and tissue samples. Conducting a retrospective analysis of data from previously analysed samples revealed the presence of lactoyl-CoA in diverse cell and tissue contexts. In addition, we describe a biosynthetic route to generate 13 C 3 15 N 1 -isotopically labelled lactoyl-CoA, providing a co-eluting internal standard for analysis of this metabolite. We estimate lactoyl-CoA concentrations of 1.14 × 10 −8 pmol per cell in cell culture and 0.0172 pmol mg −1 tissue wet weight in mouse heart. These levels are similar to crotonyl-CoA, but between 20 and 350 times lower than predominant acyl-CoAs such as acetyl-, propionyl- and succinyl-CoA. Overall our studies provide the first quantitative measurements of lactoyl-CoA in metazoans, and provide a methodological foundation for the interrogation of this novel metabolite in biology and disease.
- Published
- 2020
33. Author response for 'Quantification of lactoyl-CoA (lactyl-CoA) by liquid chromatography mass spectrometry in mammalian cells and tissues'
- Author
-
Sophie Trefely, Eliana von Krusenstiern, Erika L. Varner, Carmen Bekeova, Luke Izzo, Kathryn E. Wellen, Jordan L. Meier, Nathaniel W. Snyder, Erin L. Seifert, Roddy S. O’Connor, and David Bartee
- Subjects
Chromatography ,Liquid chromatography–mass spectrometry ,Chemistry - Published
- 2020
34. CAR-T cell-mediated depletion of immunosuppressive tumor-associated macrophages promotes endogenous antitumor immunity and augments adoptive immunotherapy
- Author
-
Daniel J. Powell, Roddy S. O’Connor, Nicholas G. Minutolo, Rachel C. Lynn, Lauren C. Shaw, Victoria Casado-Medrano, Mathilde Poussin, Takami Matsuyama, Alba Rodriguez-Garcia, Monika A. Eiva, and Gonzalo Lopez
- Subjects
0301 basic medicine ,Cancer microenvironment ,Science ,medicine.medical_treatment ,Cell ,General Physics and Astronomy ,Cancer immunotherapy ,CD8-Positive T-Lymphocytes ,Immunotherapy, Adoptive ,General Biochemistry, Genetics and Molecular Biology ,Monocytes ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Antigen ,Neoplasms ,Tumor-Associated Macrophages ,medicine ,Tumor Cells, Cultured ,Tumor Microenvironment ,Animals ,Humans ,Folate Receptor 2 ,Immunosuppression Therapy ,Tumor microenvironment ,Multidisciplinary ,Receptors, Chimeric Antigen ,Chemistry ,General Chemistry ,Immunotherapy ,Chimeric antigen receptor ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Tumor progression ,Folate receptor ,030220 oncology & carcinogenesis ,Mesothelin ,Cancer research ,sense organs ,CD8 - Abstract
The immunosuppressive tumor microenvironment (TME) represents a major barrier for effective immunotherapy. Tumor-associated macrophages (TAMs) are highly heterogeneous and plastic cell components of the TME which can either promote tumor progression (M2-like) or boost antitumor immunity (M1-like). Here, we demonstrate that a subset of TAMs that express folate receptor β (FRβ) possess an immunosuppressive M2-like profile. In syngeneic tumor mouse models, chimeric antigen receptor (CAR)-T cell-mediated selective elimination of FRβ+ TAMs in the TME results in an enrichment of pro-inflammatory monocytes, an influx of endogenous tumor-specific CD8+ T cells, delayed tumor progression, and prolonged survival. Preconditioning of the TME with FRβ-specific CAR-T cells also improves the effectiveness of tumor-directed anti-mesothelin CAR-T cells, while simultaneous co-administration of both CAR products does not. These results highlight the pro-tumor role of FRβ+ TAMs in the TME and the therapeutic implications of TAM-depleting agents as preparative adjuncts to conventional immunotherapies that directly target tumor antigens., Several strategies have been attempted to target immune suppressive populations in the tumor microenvironment. Here the authors show that folate receptor β-targeted CAR-T cells eliminate immunosuppressive tumor associated macrophages, promoting endogenous antitumor immune responses and adoptive T-cell therapy in pre-clinical models.
- Published
- 2020
35. 4-1BB costimulation promotes CAR T cell survival through noncanonical NF-κB signaling
- Author
-
Carl H. June, Roddy S. O’Connor, Michael C. Milone, Michael J. May, Steven M. Albelda, and Benjamin Philipson
- Subjects
Adoptive cell transfer ,T-Lymphocytes ,T cell ,Context (language use) ,Biochemistry ,Article ,CD19 ,Cell Line ,Tumor Necrosis Factor Receptor Superfamily, Member 9 ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,Receptor ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Receptors, Chimeric Antigen ,biology ,NF-kappa B ,Cell Biology ,Chimeric antigen receptor ,Cell biology ,medicine.anatomical_structure ,Cell culture ,030220 oncology & carcinogenesis ,biology.protein ,Signal transduction ,human activities ,Signal Transduction - Abstract
Clinical response to chimeric antigen receptor (CAR) T cell therapy is correlated with CAR T cell persistence, especially for CAR T cells that target CD19(+) hematologic malignancies. 4-1BB–costimulated CAR (BBζ) T cells exhibit longer persistence after adoptive transfer than do CD28-costimulated CAR (28ζ) T cells. 4-1BB signaling improves T cell persistence even in the context of 28ζ CAR activation, which indicates distinct prosurvival signals mediated by the 4-1BB cytoplasmic domain. To specifically study signal transduction by CARs, we developed a cell-free, ligand-based activation and ex vivo culture system for CD19-specific CAR T cells. We observed greater ex vivo survival and subsequent expansion of BBζ CAR T cells when compared to 28ζ CAR T cells. We showed that only BBζ CARs activated noncanonical nuclear factor κB (ncNF-κB) signaling in T cells basally and that the anti-CD19 BBζ CAR further enhanced ncNF-κB signaling after ligand engagement. Reducing ncNF-κB signaling reduced the expansion and survival of anti-CD19 BBζ T cells and was associated with a substantial increase in the abundance of the most pro-apoptotic isoforms of Bim. Although our findings do not exclude the importance of other signaling differences between BBζ and 28ζ CARs, they demonstrate the necessary and nonredundant role of ncNF-κB signaling in promoting the survival of BBζ CAR T cells, which likely underlies the engraftment persistence observed with this CAR design.
- Published
- 2020
36. Quantification of lactoyl-CoA by liquid chromatography mass spectrometry in mammalian cells and tissues
- Author
-
Jordan L. Meier, Eliana von Krusenstiern, Luke Izzo, Erin L. Seifert, Kathryn E. Wellen, Sophie Trefely, David Bartee, Erika L. Varner, Roddy S. O’Connor, Carmen Bekeova, and Nathaniel W. Snyder
- Subjects
chemistry.chemical_classification ,biology ,Chemistry ,Metabolite ,Cell ,Lysine ,Acylation ,Metabolic pathway ,chemistry.chemical_compound ,Histone ,medicine.anatomical_structure ,Enzyme ,Biochemistry ,Liquid chromatography–mass spectrometry ,biology.protein ,medicine - Abstract
Lysine lactoylation is a recently described protein post-translational modification (PTM). However, the biochemical pathways responsible for this acylation remain unclear. Two metabolite-dependent mechanisms have been proposed: enzymatic histone lysine lactoylation derived from lactoyl-coenzyme A (lactoyl-CoA, also termed lactyl-CoA), and non-enzymatic lysine lactoylation resulting from acyl-transfer via lactoyl-glutathione. While the former has precedent in the form of enzyme-catalyzed lysine acylation, the lactoyl-CoA metabolite has not been previously quantified in mammalian systems. Here we use liquid chromatography-high resolution mass spectrometry (LC-HRMS) together with a synthetic standard to detect and validate the presence of lactoyl-CoA in cell and tissue samples. Conducting a retrospective analysis of data from previously analyzed samples revealed the presence of lactoyl-CoA in diverse cell and tissue contexts. In addition, we describe a biosynthetic route to generate 13C315N1 -isotopically-labeled lactoyl-CoA, providing a co-eluting internal standard for analysis of this metabolite. We estimate lactoyl-CoA concentrations of 1.14×10−8 pmol/cell in cell culture and 0.0172 pmol/mg tissue wet weight in mouse heart. These levels are similar to crotonyl-CoA, but between 20-350 times lower than predominant acyl-CoAs such as acetyl-, propionyl-, and succinyl-CoA. Overall our studies provide the first quantitative measurements of lactoyl-CoA and provide a methodological foundation for the interrogation of this novel metabolite in biology and disease.Highlights- Detection of lactoyl-CoA at picomole concentrations across tissues and cells- Lactoyl-CoA was detected at concentrations similar to crontonyl-CoA within HepG2 cells- Isotopically labeled 13C315N1-lactoyl-CoA can be prepared by SILEC
- Published
- 2020
37. Testing the Specificity of Compounds Designed to Inhibit CPT1A in T Cells
- Author
-
Roddy S. O’Connor and Michael C. Milone
- Subjects
0301 basic medicine ,medicine.medical_treatment ,T-Lymphocytes ,Immunoblotting ,Drug Evaluation, Preclinical ,Lymphocyte Activation ,Article ,Small hairpin RNA ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Antigen ,medicine ,Humans ,Enzyme Inhibitors ,chemistry.chemical_classification ,Tumor microenvironment ,Carnitine O-Palmitoyltransferase ,Effector ,Fatty acid ,Immunotherapy ,Cell biology ,030104 developmental biology ,chemistry ,Epoxy Compounds ,Flux (metabolism) ,030217 neurology & neurosurgery ,Etomoxir - Abstract
In response to antigen and costimulation, T cells undergo a series of metabolic transitions that fulfill the biosynthetic demands of clonal expansion, differentiation, and effector function. Following antigen clearance, the oxidation of long-chain fatty acids (LCFAO) has been implicated in the transition from effector to central memory T cells. However, studies demonstrating a role for LCFAO in memory T-cell development have largely relied on the use of etomoxir (ETO), a small molecule inhibitor of the long-chain fatty acid transporter CPT1A. Understanding how the depletion of nutrients including LCFA that might occur in tumor microenvironments affects T-cell proliferation, differentiation, and function has important implications for tumor immunotherapy. Here, we combine the analysis of posttranscriptional gene silencing with extracellular flux assays to determine if etomoxir exerts nonspecific effects on oxidative metabolism. The off-target effects of ETO that we describe highlight the challenges of using pharmacologic inhibitors in loss-of-function approaches in T cells.
- Published
- 2020
38. CAR T cell immunotherapy for human cancer
- Author
-
Carl H. June, Omkar U. Kawalekar, Michael C. Milone, Saba Ghassemi, and Roddy S. O’Connor
- Subjects
0301 basic medicine ,medicine.medical_specialty ,T-Lymphocytes ,medicine.medical_treatment ,T cell ,Cell- and Tissue-Based Therapy ,Mutant Chimeric Proteins ,Receptors, Antigen, T-Cell ,Immunotherapy, Adoptive ,03 medical and health sciences ,Antigen ,Neoplasms ,medicine ,Humans ,Cell Engineering ,Clinical Trials as Topic ,Multidisciplinary ,Transfusion medicine ,Immunotherapy ,medicine.disease ,Chimeric antigen receptor ,Lymphoma ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,Cancer research ,Car t cells ,Genetic Engineering - Abstract
Adoptive T cell transfer (ACT) is a new area of transfusion medicine involving the infusion of lymphocytes to mediate antitumor, antiviral, or anti-inflammatory effects. The field has rapidly advanced from a promising form of immuno-oncology in preclinical models to the recent commercial approvals of chimeric antigen receptor (CAR) T cells to treat leukemia and lymphoma. This Review describes opportunities and challenges for entering mainstream oncology that presently face the CAR T field, with a focus on the challenges that have emerged over the past several years.
- Published
- 2018
39. Improving T Cell Expansion with a Soft Touch
- Author
-
Roddy S. O’Connor, Tai-De Li, Lance C. Kam, Sarah E. De Leo, Jinglun Yuan, Lester H. Lambert, Michael C. Milone, Selene Nunez-Cruz, and Geraldine K. E. Goebrecht
- Subjects
0301 basic medicine ,Materials science ,CD3 Complex ,Cell Survival ,Surface Properties ,T-Lymphocytes ,T cell ,Bioengineering ,Nanotechnology ,02 engineering and technology ,Antibodies ,Article ,03 medical and health sciences ,CD28 Antigens ,medicine ,Humans ,Cytotoxic T cell ,General Materials Science ,Dimethylpolysiloxanes ,Particle Size ,Perforin production ,Antigen-presenting cell ,Cell Proliferation ,Mechanical Engineering ,CD28 ,General Chemistry ,Microbead (research) ,021001 nanoscience & nanotechnology ,Condensed Matter Physics ,Microspheres ,030104 developmental biology ,Cell killing ,medicine.anatomical_structure ,Biophysics ,Emulsions ,Immunotherapy ,0210 nano-technology ,CD8 ,T-Lymphocytes, Cytotoxic - Abstract
Protein-coated microbeads provide a consistent approach for activating and expanding populations of T cells for immunotherapy, but don’t fully capture the properties of antigen presenting cells. In this report, we enhance T cell expansion by replacing the conventional, rigid bead with a mechanically soft elastomer. Polydimethylsiloxane (PDMS) was prepared in a microbead format and modified with activating antibodies to CD3 and CD28. Three different formulations of PDMS provided an extended proliferative phase in both CD4+-only and mixed CD4+/CD8+ T cell preparations. CD8+ T cells retained cytotoxic function, as measured by a set of biomarkers (perforin production, LAMP2 mobilization, and IFN-γ secretion) and an in vivo assay of targeted cell killing. Notably, PDMS beads presented a nanoscale polymer structure and higher rigidity than associated with conventional bulk material. These data suggest T cells respond to this higher rigidity, indicating an unexpected effect of curing conditions. Together, these studies demonstrate that adopting mechanobiology ideas into the bead platform can provide new tools for T cell-based immunotherapy.
- Published
- 2017
40. Chimeric Antigen Receptor (CAR) T cells on demand: developing potent CAR T cells in less than 24 hr for adoptive immunotherapy
- Author
-
Selene Nunez-Cruz, Saba Ghassemi, John Leferovich, Roddy S. O’Connor, Michael C. Milone, and Jai Patel
- Subjects
Cancer Research ,Transplantation ,biology ,medicine.medical_treatment ,T cell ,CD3 ,Immunology ,T-cell receptor ,CD28 ,Cell Biology ,Immunotherapy ,Gene delivery ,Chimeric antigen receptor ,medicine.anatomical_structure ,Oncology ,medicine ,Cancer research ,biology.protein ,Immunology and Allergy ,Cytotoxic T cell ,Genetics (clinical) - Abstract
Background & Aim The recent success of immunotherapy using chimeric antigen receptor modified T cells (CAR T) in B-cell malignancies highlights the potential of these cytotoxic “drugs” for cancer therapy. CAR T therapies generally relies upon viral transduction of T cell-receptor (TCR) activated T cell followed by ex vivo expansion of patient-derived T cells over days to weeks prior to infusion. In addition to the required time and labor, we previously reported that TCR/CD3 activation and ex vivo expansion leads to progressive differentiation of the CAR T cells with associated loss of CAR T cell potency. Since cell division is not a prerequisite for lentiviral vector-mediated gene delivery, we hypothesized that elimination of the CD3/CD28 activation step would yield a T cell product with high functional potency. Methods, Results & Conclusion Here, we show that functional CD19-specific CAR T cells (CART19) can be generated in as little as 24 hours using lentiviral vectors without the need for prior T cell activation. We further demonstrate that some of the restriction factors that limit the infection of quiescent T cells can be overcome by changes to the medium formulation. Most importantly, we show that non-activated T cells exhibit potent antitumor function with deep and durable control of leukemia achieved in vivo with greater than 1-log fewer CD19-specific CAR T cells compared with a 9-day process currently used by tisagenlecleucel (Fig. 1). These findings confirmed that as few as an estimated 12,000-32,000 non-activated CAR T cells produced within 24 hours from mononuclear cell collection could eradicate leukemia. These results demonstrate the potential for vastly reducing the time, materials and labor required to generate CAR T cells, which could be especially beneficial in patients with rapidly progressive disease and in resource-poor health care environments.
- Published
- 2020
41. Author Correction: Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia
- Author
-
Iulian Pruteanu-Malinici, Alina C. Boesteanu, Noelle V. Frey, Felipe Bedoya, Jennifer Brogdon, Don L. Siegel, Changfeng Zhang, F. Brad Johnson, Megan M. Davis, Edward Pequignot, Regina M. Young, Simon F. Lacey, Yan Wang, David L. Porter, Roddy S. O’Connor, Carl H. June, Sadik H. Kassim, Wei-Ting Hwang, David E Ambrose, Lifeng Tian, Joseph A. Fraietta, Corin L. Dorfmeier, Stefan Lundh, Bruce L. Levine, Jun Xu, Hans Bitter, Fang Chen, Irina Kulikovskaya, Nicholas Wilcox, Mercy Gohil, Harit Parakandi, Alexander C. Huang, E. John Wherry, Li Liu, J. Joseph Melenhorst, Elena Orlando, and Minnal Gupta
- Subjects
biology ,business.industry ,Chronic lymphocytic leukemia ,General Medicine ,medicine.disease ,General Biochemistry, Genetics and Molecular Biology ,Chimeric antigen receptor ,CD19 ,Text mining ,Cancer research ,biology.protein ,Medicine ,CAR T-cell therapy ,business - Published
- 2021
42. Novel media formulations to enhance Chimeric Antigen Receptor (CAR) T-cell potency and anti-tumor cell function for adoptive immunotherapy
- Author
-
David Heo, Sarah A. Richman, John Leferovich, Yunqi Lu, Saba Ghassemi, Alyssa Master, Juan Carlos García-Cañaveras, Y. Tu, A. Wang, Michael C. Milone, Asma Ayari, Roddy S. O’Connor, Francisco J. Martinez-Becerra, and Joshua D. Rabinowitz
- Subjects
Cancer Research ,Transplantation ,Growth factor ,medicine.medical_treatment ,T cell ,Immunology ,Cell Biology ,Biology ,Gene delivery ,Chimeric antigen receptor ,Blood cell ,medicine.anatomical_structure ,Oncology ,Cell culture ,medicine ,Cancer research ,Immunology and Allergy ,Genetics (clinical) ,Ex vivo ,Fetal bovine serum - Abstract
Background & Aim Effective CAR T-cell therapy is dependent on optimal cell culture methods conducive to the activation and expansion of T cells ex vivo as well as infection with CAR. In this regard, the outcome of the ex-vivo process is largely dependent on the properties of the medium. Media formulations used in CAR T-cell manufacturing have not been optimized either for signaling studies, gene delivery or cell expansion. Bioactive components and derivatives that support the generation of functionally-competent T cell progeny with long-lasting persistence are largely undefined. Current media formulations rely on fetal bovine serum (FBS) or human serum (HS) which suffer from a lack of consistency or supply issues. We recognize that components of blood cellular fractions that are absent in serum may have therapeutic value. Here we investigate if a concentrated growth factor extract, purified from human transfusion grade whole blood fractions, and marketed as PhysiologixTM XF hGFC (Phx), supports CAR T-cell expansion and function. Methods, Results & Conclusion We show that Phx supports T cell proliferation in clinical as well as research-grade media. As cytokine stimulation regulates the exo and endocytotic machinery, we hypothesized that factors unique to the growth factors within blood cell fractions would enhance the dynamic process of viral entry. We show that Phx treatment enhances lentiviral-mediated gene expression across a wide range of multiplicity of infections (MOIs). We compared the ability of anti-GD-2 CAR T-cells expanded ex vivo in medium conditioned with either Phx or HS to clear tumor burden in a human xenograft model of neuroblastoma. We show that CAR T-cells expanded in Phx have superior engraftment and potency in vivo. Metabolomic profiling revealed several factors enriched in a blood cell-derived growth factor concentrate that may enhance lentiviral-mediated gene delivery and CAR T-cell differentiation for preclinical discovery, process development, as well as manufacturing. These findings are also informative regarding potential derivatives to include in medium customized for gene delivery, T cell activation, and expansion for adoptive immunotherapies.
- Published
- 2020
43. Abstract PR07: Human chimeric antigen receptor (CAR) macrophages for cancer immunotherapy
- Author
-
Miroslaw Kozlowski, Kristin Blouch, Roddy S. O’Connor, Benjamin A. Garcia, Andrew Best, Michael Klichinsky, Stephen R. Wallace, Saar Gill, Dylan M. Marchione, Maksim Shestov, Xueqing Maggie Lu, Miriam Y. Kim, Olga Shestova, Carl H. June, Marco Ruella, and Saad S. Kenderian
- Subjects
Cancer Research ,Tumor microenvironment ,business.industry ,medicine.medical_treatment ,Immunology ,Immunotherapy ,Chimeric antigen receptor ,Cell therapy ,Immune system ,Cancer immunotherapy ,Antigen ,Interferon ,Cancer research ,Medicine ,business ,medicine.drug - Abstract
Despite recent landmark advances in T-cell immunotherapy for the treatment of human cancer, metastatic solid tumors remain an intractable challenge. Macrophages are often the most abundant immune cell in the tumor microenvironment (TME), where they may convert into immunosuppressive (M2) tumor-associated macrophages (TAMs) and participate in disease progression. Currently, macrophage-orientated immunotherapeutic approaches under clinical development in oncology seek to reduce TAM infiltration (CSF-1 antagonists) or enhance TAM phagocytosis (CD47 antagonists). Transfer of autologous, activated, but nontargeted macrophages failed to demonstrate antitumor efficacy in past clinical trials. We hypothesized that genetically engineering human macrophages with CARs against tumor-associated antigens could redirect their phagocytic activity and lead to therapeutic efficacy with the potential for the induction of an antitumor T-cell response. We first demonstrate that CD3-zeta-based CARs are capable of inducing phagocytosis in human THP-1 macrophages, while truncated intracellular-domain deficient CARs were not. Targeted phagocytosis and clearance of CD19+, mesothelin +, and HER2+ cells by CARs targeted against each respective antigen was significantly superior to that by control untransduced (UTD) macrophages. We demonstrate that primary human macrophages, which are resistant to most viral vectors, are efficiently transduced by the chimeric fiber adenoviral vector Ad5f35 (~70% in 10 normal donors). Using Ad5f35, we engineered primary human macrophages with a CD3-zeta-based CAR against HER2. Anti-HER2 primary human CAR macrophages demonstrated targeted phagocytosis against HER2+ but not HER2- cell lines, with phagocytic activity dependent on both the CAR and antigen densities. Furthermore, CAR, but not UTD, macrophages led to potent dose-dependent killing of three distinct HER2-high cell lines in vitro. We sought to test the efficacy of anti-HER2 primary human macrophages in xenograft models of human HER2+ ovarian cancer. A single dose of CAR, but not UTD macrophages, led to tumor regression and improved overall survival in both intraperitoneal and disseminated models of disease. We show that macrophage transduction with Ad5f35, a double-stranded DNA virus, leads to a broad gene expression change, an interferon signaling signature, and phenotypic clustering toward classically activated M1 macrophages. CAR macrophages upregulated co-stimulatory ligand and antigen processing/presentation genes and led to enhanced T-cell stimulation in vitro and in vivo. Lastly, CAR, but not UTD, macrophages showed a broad resistance for M2 conversion in response to immunosuppressive cytokines. In conclusion, we show that primary human CAR macrophages are capable of targeted tumor phagocytosis, lead to improved overall survival in xenograft models, and demonstrate enhanced T-cell stimulation. CAR macrophages are a novel cell therapy platform for the treatment of human cancer. This abstract is also being presented as Poster B29. Citation Format: Michael Klichinsky, Marco Ruella, Olga Shestova, Andrew Best, Kristin Blouch, Xueqing M. Lu, Saad S. Kenderian, Miriam Y. Kim, Roddy O'Connor, Stephen Wallace, Miroslaw Kozlowski, Dylan M. Marchione, Maksim Shestov, Benjamin A. Garcia, Carl June, Saar Gill. Human chimeric antigen receptor (CAR) macrophages for cancer immunotherapy [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2018 Nov 27-30; Miami Beach, FL. Philadelphia (PA): AACR; Cancer Immunol Res 2020;8(4 Suppl):Abstract nr PR07.
- Published
- 2020
44. Reducing Ex Vivo Culture Improves the Antileukemic Activity of Chimeric Antigen Receptor (CAR) T Cells
- Author
-
Prachi R. Patel, Megan M. Davis, Joseph A. Fraietta, David M. Barrett, Simon F. Lacey, John Scholler, John Leferovich, Selene Nunez-Cruz, Stephan A. Grupp, Felipe Bedoya, J. Joseph Melenhorst, Michael C. Milone, Saba Ghassemi, Bruce L. Levine, Roddy S. O’Connor, Stefan Lundh, Changfeng Zhang, and Carl H. June
- Subjects
0301 basic medicine ,Cytotoxicity, Immunologic ,Cancer Research ,Adoptive cell transfer ,Time Factors ,medicine.medical_treatment ,T-Lymphocytes ,Immunology ,Antigens, CD19 ,Cell Culture Techniques ,Lymphocyte Activation ,Immunotherapy, Adoptive ,CD19 ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Antigen ,Antigens, Neoplasm ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Receptors, Chimeric Antigen ,biology ,business.industry ,Cell Differentiation ,Immunotherapy ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Xenograft Model Antitumor Assays ,In vitro ,Chimeric antigen receptor ,Leukemia ,030104 developmental biology ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,business ,Ex vivo - Abstract
The success of chimeric antigen receptor (CAR)–mediated immunotherapy in acute lymphoblastic leukemia (ALL) highlights the potential of T-cell therapies with directed cytotoxicity against specific tumor antigens. The efficacy of CAR T-cell therapy depends on the engraftment and persistence of T cells following adoptive transfer. Most protocols for T-cell engineering routinely expand T cells ex vivo for 9 to 14 days. Because the potential for engraftment and persistence is related to the state of T-cell differentiation, we hypothesized that reducing the duration of ex vivo culture would limit differentiation and enhance the efficacy of CAR T-cell therapy. We demonstrated that T cells with a CAR-targeting CD19 (CART19) exhibited less differentiation and enhanced effector function in vitro when harvested from cultures at earlier (day 3 or 5) compared with later (day 9) timepoints. We then compared the therapeutic potential of early versus late harvested CART19 in a murine xenograft model of ALL and showed that the antileukemic activity inversely correlated with ex vivo culture time: day 3 harvested cells showed robust tumor control despite using a 6-fold lower dose of CART19, whereas day 9 cells failed to control leukemia at limited cell doses. We also demonstrated the feasibility of an abbreviated culture in a large-scale current good manufacturing practice–compliant process. Limiting the interval between T-cell isolation and CAR treatment is critical for patients with rapidly progressing disease. Generating CAR T cells in less time also improves potency, which is central to the effectiveness of these therapies. Cancer Immunol Res; 6(9); 1100–9. ©2018 AACR.
- Published
- 2018
45. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia
- Author
-
Lifeng Tian, Joseph A. Fraietta, Jun Xu, Hans Bitter, Carl H. June, Iulian Pruteanu-Malinici, Edward Pequignot, Alina C. Boesteanu, Roddy S. O’Connor, Changfeng Zhang, Noelle V. Frey, Corin L. Dorfmeier, F. Brad Johnson, Simon F. Lacey, Nicholas Wilcox, Bruce L. Levine, Sadik H. Kassim, Stefan Lundh, Harit Parakandi, E. John Wherry, Megan M. Davis, J. Joseph Melenhorst, Regina M. Young, David L. Porter, Wei-Ting Hwang, Yan Wang, Felipe Bedoya, Alexander C. Huang, David E Ambrose, Li Liu, Mercy Gohil, Jennifer Brogdon, Elena Orlando, Don L. Siegel, Fang Chen, Irina Kulikovskaya, and Minnal Gupta
- Subjects
0301 basic medicine ,Male ,STAT3 Transcription Factor ,Transcription, Genetic ,medicine.medical_treatment ,Chronic lymphocytic leukemia ,T-Lymphocytes ,Antigens, CD19 ,Cell- and Tissue-Based Therapy ,Biology ,Immunotherapy, Adoptive ,General Biochemistry, Genetics and Molecular Biology ,CD19 ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Antigen ,medicine ,Cytotoxic T cell ,Animals ,Humans ,Receptors, Chimeric Antigen ,Interleukin-6 ,General Medicine ,Immunotherapy ,medicine.disease ,Leukemia, Lymphocytic, Chronic, B-Cell ,Chimeric antigen receptor ,Leukemia ,030104 developmental biology ,Treatment Outcome ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Female - Abstract
Tolerance to self-antigens prevents the elimination of cancer by the immune system1,2. We used synthetic chimeric antigen receptors (CARs) to overcome immunological tolerance and mediate tumor rejection in patients with chronic lymphocytic leukemia (CLL). Remission was induced in a subset of subjects, but most did not respond. Comprehensive assessment of patient-derived CAR T cells to identify mechanisms of therapeutic success and failure has not been explored. We performed genomic, phenotypic and functional evaluations to identify determinants of response. Transcriptomic profiling revealed that CAR T cells from complete-responding patients with CLL were enriched in memory-related genes, including IL-6/STAT3 signatures, whereas T cells from nonresponders upregulated programs involved in effector differentiation, glycolysis, exhaustion and apoptosis. Sustained remission was associated with an elevated frequency of CD27+CD45RO–CD8+ T cells before CAR T cell generation, and these lymphocytes possessed memory-like characteristics. Highly functional CAR T cells from patients produced STAT3-related cytokines, and serum IL-6 correlated with CAR T cell expansion. IL-6/STAT3 blockade diminished CAR T cell proliferation. Furthermore, a mechanistically relevant population of CD27+PD-1–CD8+ CAR T cells expressing high levels of the IL-6 receptor predicts therapeutic response and is responsible for tumor control. These findings uncover new features of CAR T cell biology and underscore the potential of using pretreatment biomarkers of response to advance immunotherapies. An IL-6/STAT3 signature and memory CD8 T cell subset in preinfusion chimeric antigen receptor–expressing T cells associate with response in patients with high-risk chronic lymphocytic leukemia.
- Published
- 2018
46. The CPT1a inhibitor, etomoxir induces severe oxidative stress at commonly used concentrations
- Author
-
Saba Ghassemi, Lili Guo, Nathaniel W. Snyder, Yoonseok Kam, Liwei Weng, Carl H. June, Michael C. Milone, Ian A. Blair, Sophie Trefely, Andrew J. Worth, Roddy S. O’Connor, Benjamin Philipson, and Selene Nunez-Cruz
- Subjects
0301 basic medicine ,T cell ,T-Lymphocytes ,lcsh:Medicine ,Pharmacology ,medicine.disease_cause ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,medicine ,Humans ,Carnitine ,Enzyme Inhibitors ,lcsh:Science ,Beta oxidation ,Cell Proliferation ,Multidisciplinary ,Carnitine O-Palmitoyltransferase ,Dose-Response Relationship, Drug ,Chemistry ,Cell growth ,lcsh:R ,CD28 ,Glutamine ,Oxidative Stress ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Epoxy Compounds ,lcsh:Q ,Etomoxir ,Oxidative stress ,medicine.drug - Abstract
Etomoxir (ETO) is a widely used small-molecule inhibitor of fatty acid oxidation (FAO) through its irreversible inhibitory effects on the carnitine palmitoyl-transferase 1a (CPT1a). We used this compound to evaluate the role of fatty acid oxidation in rapidly proliferating T cells following costimulation through the CD28 receptor. We show that ETO has a moderate effect on T cell proliferation with no observable effect on memory differentiation, but a marked effect on oxidative metabolism. We show that this oxidative metabolism is primarily dependent upon glutamine rather than FAO. Using an shRNA approach to reduce CPT1a in T cells, we further demonstrate that the inhibition of oxidative metabolism in T cells by ETO is independent of its effects on FAO at concentrations exceeding 5 μM. Concentrations of ETO above 5 μM induce acute production of ROS with associated evidence of severe oxidative stress in proliferating T cells. In aggregate, these data indicate that ETO lacks specificity for CTP1a above 5 μM, and caution should be used when employing this compound for studies in cells due to its non-specific effects on oxidative metabolism and cellular redox.
- Published
- 2018
47. Simple, 1-Day Manufacturing of Quiescent Chimeric Antigen Receptor T Cells for Adoptive Immunotherapy
- Author
-
Jai Patel, Selene Nunez-Cruz, Roddy S. O’Connor, Saba Ghassemi, Michael C. Milone, and John Leferovich
- Subjects
medicine.medical_treatment ,Immunology ,Adoptive immunotherapy ,Cancer therapy ,Cell Biology ,Hematology ,Immunotherapy ,Biology ,medicine.disease ,Biochemistry ,Chimeric antigen receptor ,Leukemia ,Cytokine ,Antigen ,Acute lymphocytic leukemia ,Cancer research ,medicine - Abstract
The recent success of immunotherapy using chimeric antigen receptor modified T cells (CAR T) in B-cell malignancies highlights the potential of these cytotoxic "drugs" for cancer therapy. CAR T therapies generally rely upon manufacturing approaches that include prior T cell activation through engagement of the TCR and costimulatory receptors followed by ex vivo expansion of patient-derived T cells over days to weeks. We previously reported that CD3/CD28 stimulation prior to transduction promotes progressive T cell effector differentiation over time in culture with loss of CAR T cell potency (Ghassemi et al. 2018 PMID: 30030295). Since cell division is not a prerequisite for lentiviral vector-mediated gene delivery, we hypothesized that lentiviral transduction of quiescent T cells without prior activation will enhance engraftment and persistence of CART cells that is associated with long-term leukemia control. Here, we show that functional CD19-specific CAR T cells (CART19) can be generated in as little as 24 hours using lentiviral vectors without the need for prior T cell activation. We showed using a non-optimized process that a mean of 6.5% (range 2%-10%) of freshly isolated quiescent T cells can be transduced using an infrared red fluorescent protein (iRFP)-expressing lentiviral vector with slower kinetics of expression compared with activated T cell transduction (peak at 96 hrs vs. 48 hrs for quiescent and activated T cells, respectively). Although substantially less efficient compared to activated T cells, transduction was detected across all T cells subsets with central memory T cells showing the greatest transduction efficiency with a mean of 4-fold greater transduction compared with naïve T cells. Somewhat unexpectedly, CART19 cells generated from quiescent T cells using a CD19-specific CAR vector showed a 3-5 fold greater transgene expression compared with iRFP vectors transduced at similar MOI. However, we show that CAR expression can occur in quiescent T cells even without reverse transcription or integrase function, so called "pseudotransduction". Importantly, we show that this CAR expression produces T cells with cytolytic activity and effector cytokine production in response to antigen that is similar to activated and transduced CAR T cells. Using the well-characterized Nalm6 model of acute lymphoblastic leukemia, we show that CART19 cells generated by transduction of quiescent T cells for 16 hours followed by washing to remove vector exhibit dose-dependent anti-leukemic activity that is durable with injection of as little as 2x105 total T cells. We estimate the latter to contain ~2x104 T cells with integrated lentiviral vector based upon transduction efficiency determined in studies using non-tumor bearing mice. (Fig 1). In summary, our results support the need for further investigation of CAR T cells that are generated using engineering of quiescent T cells. Taking advantage of the ability of lentiviral vectors to transfer genes to quiescent T cells, the highly abbreviated and simplified manufacturing approach described here has the potential to enhance therapeutic potency while also substantially reducing the materials and labor costs associated with current manufacturing approaches that use activated and expanded T cells. In addition, the rapid nature of this manufacturing has the potential to extend the population of patients that may be treated with these therapies by shortening the interval between aphaeresis collection and re-infusion of CAR T cells, which prevents the treatment of some patients with rapidly progressive disease. Disclosures Ghassemi: Novartis: Patents & Royalties. Milone:Novartis: Patents & Royalties, Research Funding.
- Published
- 2019
48. Abstract B029: A novel media supplementation strategy for improved T-cell culture and preservation of naivety
- Author
-
Alyssa Master, Lisa Karimi-Naser, Saba Ghassemi, Roddy S. O’Connor, and Dina A. Schneider
- Subjects
Cancer Research ,education.field_of_study ,Naivety ,T cell ,medicine.medical_treatment ,Immunology ,Cell ,Population ,CD28 ,Biology ,Andrology ,Transduction (genetics) ,medicine.anatomical_structure ,Cancer immunotherapy ,medicine ,education ,Fetal bovine serum - Abstract
Recent advances in genetic engineering have resulted in exponential growth in cell therapy technologies. As an increasing number of CAR-T therapies are entering clinical trials, there is a scarcity of resources put into optimizing production strategies. This is largely due to the highly competitive timelines for these companies to advance their drug candidate. Yet, due to this pressure, there is a real need for advances in the cell culture media used to recover, sustain and expand these importanT-cells. Here we show that a novel xeno-free media supplement, PhysiologixTM XF serum replacement, results in improved human primary T-cell growth, preservation of naïve and central memory phenotype, and enhanced transduction efficiency when used in place of traditional media supplements. In the presence of this supplement, T-cell proliferation was nearly doubled in comparison to industry-leading serum free media. In a separate study, we compared PhysiologixTM XF to a control media containing the traditional 10% fetal bovine serum for bulk T-cell culture. We observed that T-cells in both control and test media had similar cell size and population doublings following 48 hours of CD3/CD28 activation. However, FACS analysis revealed thaT-cells grown in PhysiologixTM XF showed a 33% increase in beneficial naïve-like and central memory T-cell phenotypes after nine days in culture when compared to control media. Additionally, lentiviral transduction efficiency was increased from 74% to 94% in PhysiologixTM XF supplemented cells compared to control cells. Taken together, these data demonstrate that T-cell culture with PhysiologixTM XF results in improved growth characteristics, desirable cell phenotypes for patient infusion, and improved ease of transduction, all of which may prove beneficial in the CAR-T race from bench to bedside. Citation Format: Alyssa Master, Roddy S. O'Connor, Saba Ghassemi, Dina A. Schneider, Lisa Karimi-Naser. A novel media supplementation strategy for improved T-cell culture and preservation of naivety [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr B029.
- Published
- 2019
49. Programmed death ligand-1 expression on donor T cells drives graft-versus-host disease lethality
- Author
-
Arlene H. Sharpe, Ian A. Blair, William J. Murphy, David H. Munn, Roddy S. O’Connor, Jonathan S. Serody, Catarina Sacristán, Govindarajan Thangavelu, Caleph B. Wilson, Gordon J. Freeman, Jan M. Pawlicki, Michael C. Milone, Bruce R. Blazar, Scott B. Lovitch, Asim Saha, Durga Bhavani Dandamudi, Rocio Foncea, James L. Riley, Laurence A. Turka, Kazutoshi Aoyama, Scott N. Furlan, Parvathi Ranganathan, Patricia A. Taylor, Brian T. Fife, David A. Bernlohr, Angela Panoskaltsis-Mortari, Benjamin G. Vincent, Leslie S. Kean, Joel S. Burrill, Lili Guo, Steven M. Devine, Victor Tkachev, Michael L. Dustin, Jeffrey S. Miller, and Nathaniel W. Snyder
- Subjects
0301 basic medicine ,T-Lymphocytes ,Glutamine ,Programmed Cell Death 1 Receptor ,Graft vs Host Disease ,Apoptosis ,Inbred C57BL ,Medical and Health Sciences ,B7-H1 Antigen ,Interleukin 21 ,Mice ,0302 clinical medicine ,Leukocytes ,Cytotoxic T cell ,2.1 Biological and endogenous factors ,IL-2 receptor ,Phosphorylation ,Aetiology ,Inbred BALB C ,Bone Marrow Transplantation ,Cancer ,Mice, Inbred BALB C ,General Medicine ,medicine.anatomical_structure ,Treatment Outcome ,Cytokines ,Female ,Stem Cell Research - Nonembryonic - Non-Human ,medicine.symptom ,Glycolysis ,Research Article ,Signal Transduction ,T cell ,Mononuclear ,Immunology ,Inflammation ,Bone Marrow Cells ,Biology ,03 medical and health sciences ,Immune system ,Rare Diseases ,medicine ,Animals ,Humans ,Transplantation ,Inflammatory and immune system ,medicine.disease ,Stem Cell Research ,Mice, Inbred C57BL ,Oxygen ,030104 developmental biology ,Graft-versus-host disease ,Glucose ,Orphan Drug ,Leukocytes, Mononuclear ,030215 immunology - Abstract
Programmed death ligand-1 (PD-L1) interaction with PD-1 induces T cell exhaustion and is a therapeutic target to enhance immune responses against cancer and chronic infections. In murine bone marrow transplant models, PD-L1 expression on host target tissues reduces the incidence of graft-versus-host disease (GVHD). PD-L1 is also expressed on T cells; however, it is unclear whether PD-L1 on this population influences immune function. Here, we examined the effects of PD-L1 modulation of T cell function in GVHD. In patients with severe GVHD, PD-L1 expression was increased on donor T cells. Compared with mice that received WT T cells, GVHD was reduced in animals that received T cells from Pdl1-/- donors. PD-L1–deficient T cells had reduced expression of gut homing receptors, diminished production of inflammatory cytokines, and enhanced rates of apoptosis. Moreover, multiple bioenergetic pathways, including aerobic glycolysis, oxidative phosphorylation, and fatty acid metabolism, were also reduced in T cells lacking PD-L1. Finally, the reduction of acute GVHD lethality in mice that received Pdl1-/- donor cells did not affect graft-versus-leukemia responses. These data demonstrate that PD-L1 selectively enhances T cell–mediated immune responses, suggesting a context-dependent function of the PD-1/PD-L1 axis, and suggest selective inhibition of PD-L1 on donor T cells as a potential strategy to prevent or ameliorate GVHD.
- Published
- 2016
50. Substrate Rigidity Regulates Human T Cell Activation and Proliferation
- Author
-
Xueli Hao, Tatiana Akimova, Lance C. Kam, Michael C. Milone, Roddy S. O’Connor, Wayne W. Hancock, Keenan T. Bashour, and Keyue Shen
- Subjects
biology ,Chemistry ,Cell growth ,T cell ,CD3 ,Immunology ,T-cell receptor ,CD28 ,medicine.anatomical_structure ,Biochemistry ,Cell culture ,medicine ,Biophysics ,biology.protein ,Immunology and Allergy ,Stem cell ,CD8 - Abstract
Adoptive immunotherapy using cultured T cells holds promise for the treatment of cancer and infectious disease. Ligands immobilized on surfaces fabricated from hard materials such as polystyrene plastic are commonly employed for T cell culture. The mechanical properties of a culture surface can influence the adhesion, proliferation, and differentiation of stem cells and fibroblasts. We therefore explored the impact of culture substrate stiffness on the ex vivo activation and expansion of human T cells. We describe a simple system for the stimulation of the TCR/CD3 complex and the CD28 receptor using substrates with variable rigidity manufactured from poly(dimethylsiloxane), a biocompatible silicone elastomer. We show that softer (Young’s Modulus [E] < 100 kPa) substrates stimulate an average 4-fold greater IL-2 production and ex vivo proliferation of human CD4+ and CD8+ T cells compared with stiffer substrates (E > 2 MPa). Mixed peripheral blood T cells cultured on the stiffer substrates also demonstrate a trend (nonsignificant) toward a greater proportion of CD62Lneg, effector-differentiated CD4+ and CD8+ T cells. Naive CD4+ T cells expanded on softer substrates yield an average 3-fold greater proportion of IFN-γ–producing Th1-like cells. These results reveal that the rigidity of the substrate used to immobilize T cell stimulatory ligands is an important and previously unrecognized parameter influencing T cell activation, proliferation, and Th differentiation. Substrate rigidity should therefore be a consideration in the development of T cell culture systems as well as when interpreting results of T cell activation based upon solid-phase immobilization of TCR/CD3 and CD28 ligands.
- Published
- 2012
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.