133 results on '"Berrien-Elliott, Melissa"'
Search Results
102. Human cytokine-induced memory-like NK cells expand in patients with AML and display enhanced anti-leukemia responses.
- Author
-
Berrien-Elliott, Melissa M, primary, Romee, Rizwan, additional, Wagner, Julia, additional, Rosario, Maximillian, additional, Jewell, Brea, additional, Schappe, Timothy, additional, Leong, Jeffery, additional, Abdel-Latif, Sara, additional, Willey, Sarah, additional, Yu, Liyang, additional, Oh, Stephen, additional, mulder, Arend, additional, Class, Frans, additional, and Fehniger, Todd A., additional
- Published
- 2016
- Full Text
- View/download PDF
103. Mechanisms governing human memory-like natural killer cell function.
- Author
-
Wagner, Julia A, primary, Berrien-Elliott, Melissa M, additional, Rosario, Maximillian, additional, Jewell, Brea A, additional, Leong, Jeffrey W, additional, Schappe, Timothy K, additional, Abdel-Latif, Sara, additional, and Fehniger, Todd A, additional
- Published
- 2016
- Full Text
- View/download PDF
104. Human Cytokine-Induced Memory-Like Natural Killer Cells
- Author
-
Berrien-Elliott, Melissa M., Wagner, Julia A., and Fehniger, Todd A.
- Abstract
Natural killer (NK) cells are innate lymphoid cells that are important for host defense against infection and mediate antitumor responses. Recent reports from several laboratories have identified that NK cells can remember a prior activation event and consequently respond more robustly when restimulated, a property termed innate NK cell memory. NK cell memory has now been identified following hapten exposure, viral infection, and combined cytokine preactivation with IL-12, IL-15, and IL-18. Many questions in the field remain regarding the cellular and molecular mechanisms regulating memory NK cells and their responses, as well as their formation and function in mice and humans. Here we review our current understanding of cytokine-induced memory-like (CIML) NK cells that are generated by combined preactivation with IL-12, IL-15, and IL-18. These cells exhibit enhanced NK cell effector functions weeks after the initial cytokine preactivation. Further, we highlight the preclinical rationale and ongoing therapeutic application of CIML NK cells for adoptive immunotherapy in patients with hematologic malignancies. © 2015 S. Karger AG, Basel
- Published
- 2024
- Full Text
- View/download PDF
105. Human Cytokine-Induced Memory-like NK Cells Exhibit in Vivo Anti-Leukemia Activity in Xenografted NSG Mice and in Patients with Acute Myeloid Leukemia (AML)
- Author
-
Romee, Rizwan, primary, Maximillian, Rosario, additional, Berrien-Elliott, Melissa M, additional, Wagner, Julia A, additional, Jewell, Brea A, additional, Schappe, Timothy, additional, Leong, Jeffrey W, additional, Schneider, Stephanie E, additional, Willey, Sarah, additional, and Fehniger, Todd A, additional
- Published
- 2015
- Full Text
- View/download PDF
106. Recurrent Somatic Genomic Alterations in Follicular NHL (FL) Revealed By Exome and Custom-Capture Next Generation Sequencing
- Author
-
Fehniger, Todd A, primary, Krysiak, Kilannin, additional, White, Brian S, additional, Matlock, Matthew, additional, Miller, Chris, additional, Fulton, Robert, additional, Kreisel, Friederike, additional, Fronick, Catrina, additional, Cook, Lisa, additional, Veizer, Joelle, additional, Cashen, Amanda F., additional, Carson, Kenneth R, additional, Berrien-Elliott, Melissa M, additional, Bartlett, Nancy L, additional, Wilson, Richard K, additional, Mardis, Elaine R, additional, Ley, Timothy J, additional, Griffith, Malachi, additional, and Griffith, Obi L, additional
- Published
- 2015
- Full Text
- View/download PDF
107. Improving natural killer cell cancer immunotherapy
- Author
-
Berrien-Elliott, Melissa M., primary, Romee, Rizwan, additional, and Fehniger, Todd A., additional
- Published
- 2015
- Full Text
- View/download PDF
108. MicroRNA-15/16 Antagonizes Myb To Control NK Cell Maturation
- Author
-
Sullivan, Ryan P., primary, Leong, Jeffrey W., additional, Schneider, Stephanie E., additional, Ireland, Aaron R., additional, Berrien-Elliott, Melissa M., additional, Singh, Anvita, additional, Schappe, Timothy, additional, Jewell, Brea A., additional, Sexl, Veronika, additional, and Fehniger, Todd A., additional
- Published
- 2015
- Full Text
- View/download PDF
109. First-in-Human Clinical Trial of the IL-15 Super-Agonist Complex ALT-803 for Patients Relapsed after Allogeneic Hematopoietic Cell Transplantation
- Author
-
Romee, Rizwan, Cooley, Sarah, Berrien-Elliott, Melissa, Westervelt, Peter, Verneris, Michael R, Wagner, John E., Weisdorf, Daniel J., Blazar, Bruce, DiPersio, John F, Cashen, Amanda F., Jeng, Emily K, Rock, Amy, Wong, Hing C, Fehniger, Todd A., and Miller, Jeffrey S.
- Published
- 2017
- Full Text
- View/download PDF
110. NKG2A Represents an Important Immune Checkpoint for Human Cytokine-Induced Memory-like NK Cells in Patients with AML
- Author
-
Berrien-Elliott, Melissa, Romee, Rizwan, Wagner, Julia Alexandra, Becker-Hapak, Michelle, Neal, Carly, Schappe, Tim, McCullen, Matthew, Cooper, Megan, and Fehniger, Todd A.
- Published
- 2017
- Full Text
- View/download PDF
111. Checkpoint Blockade Immunotherapy Relies on T-bet but Not Eomes to Induce Effector Function in Tumor-Infiltrating CD8+ T Cells
- Author
-
Berrien-Elliott, Melissa M., primary, Yuan, Jinyun, additional, Swier, Lauryn E., additional, Jackson, Stephanie R., additional, Chen, Collin L., additional, Donlin, Maureen J., additional, and Teague, Ryan M., additional
- Published
- 2015
- Full Text
- View/download PDF
112. Human Cytokine-Induced Memory-Like Natural Killer Cells
- Author
-
Berrien-Elliott, Melissa M., primary, Wagner, Julia A., additional, and Fehniger, Todd A., additional
- Published
- 2015
- Full Text
- View/download PDF
113. Neuropilin-1 Expression Is Induced on Tolerant Self-Reactive CD8+ T Cells but Is Dispensable for the Tolerant Phenotype
- Author
-
Jackson, Stephanie R., primary, Berrien-Elliott, Melissa, additional, Yuan, Jinyun, additional, Hsueh, Eddy C., additional, and Teague, Ryan M., additional
- Published
- 2014
- Full Text
- View/download PDF
114. Inflammation programs self-reactive CD8+ T cells to acquire T-box-mediated effector function but does not prevent deletional tolerance
- Author
-
Jackson, Stephanie R, primary, Yuan, Jinyun, additional, Berrien-Elliott, Melissa M, additional, Chen, Collin L, additional, Meyer, Jennifer M, additional, Donlin, Maureen J, additional, and Teague, Ryan M, additional
- Published
- 2014
- Full Text
- View/download PDF
115. T-bet dictates CD8+ T cell tolerance versus immunity following antigen recognition (P4368)
- Author
-
Jackson, Stephanie, primary, Yuan, Jinyun, additional, Berrien-Elliott, Melissa, additional, Meyer, Jennifer, additional, and Teague, Ryan, additional
- Published
- 2013
- Full Text
- View/download PDF
116. CD8+T Cell Exhaustion During Persistent Viral Infection is Regulated Independently of the Virus-Specific T Cell Receptor
- Author
-
Jackson, Stephanie R., primary, Berrien-Elliott, Melissa M., additional, Meyer, Jennifer M., additional, Wherry, E. John, additional, and Teague, Ryan M., additional
- Published
- 2013
- Full Text
- View/download PDF
117. Durable Adoptive Immunotherapy for Leukemia Produced by Manipulation of Multiple Regulatory Pathways of CD8+ T-Cell Tolerance
- Author
-
Berrien-Elliott, Melissa M., primary, Jackson, Stephanie R., additional, Meyer, Jennifer M., additional, Rouskey, Craig J., additional, Nguyen, Thanh-Long M., additional, Yagita, Hideo, additional, Greenberg, Philip D., additional, DiPaolo, Richard J., additional, and Teague, Ryan M., additional
- Published
- 2013
- Full Text
- View/download PDF
118. Neuropilin-1 Expression Is Induced on Tolerant Self-Reactive CD8+ T Cells but Is Dispensable for the Tolerant Phenotype.
- Author
-
Jackson, Stephanie R., Berrien-Elliott, Melissa, Yuan, Jinyun, Hsueh, Eddy C., and Teague, Ryan M.
- Subjects
- *
NEUROPILINS , *T cells , *IMMUNITY , *PHENOTYPES , *IMMUNE system - Abstract
Establishing peripheral CD8+ T cell tolerance is vital to avoid immune mediated destruction of healthy self-tissues. However, it also poses a major impediment to tumor immunity since tumors are derived from self-tissue and often induce T cell tolerance and dysfunction. Thus, understanding the mechanisms that regulate T cell tolerance versus immunity has important implications for human health. Signals received from the tissue environment largely dictate whether responding T cells become activated or tolerant. For example, induced expression and subsequent ligation of negative regulatory receptors on the surface of self-reactive CD8+ T cells are integral in the induction of tolerance. We utilized a murine model of T cell tolerance to more completely define the molecules involved in this process. We discovered that, in addition to other known regulatory receptors, tolerant self-reactive CD8+ T cells distinctly expressed the surface receptor neuropilin-1 (Nrp1). Nrp1 was highly induced in response to self-antigen, but only modestly when the same antigen was encountered under immune conditions, suggesting a possible mechanistic link to T cell tolerance. We also observed a similar Nrp1 expression profile on human tumor infiltrating CD4+ and CD8+ T cells. Despite high expression on tolerant CD8+ T cells, our studies revealed that Nrp1 had no detectable role in the tolerant phenotype. Specifically, Nrp1-deficient T cells displayed the same functional defects as wild-type self-reactive T cells, lacking in vivo cytolytic potential, IFNγ production, and antitumor responses. While reporting mostly negative data, our findings have therapeutic implications, as Nrp1 is now being targeted for human cancer therapy in clinical trials, but the precise molecular pathways and immune cells being engaged during treatment remain incompletely defined. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
119. CD8+ T Cell Exhaustion During Persistent Viral Infection is Regulated Independently of the Virus-Specific T Cell Receptor.
- Author
-
Jackson, Stephanie R., Berrien-Elliott, Melissa M., Meyer, Jennifer M., Wherry, E. John, and Teague, Ryan M.
- Subjects
- *
CD8 antigen , *VIRUS diseases , *T cell receptors , *AUTOANTIGENS , *IMMUNIZATION , *BINDING sites , *IMMUNITY - Abstract
During chronic viral infections, responses by virus-specific CD8+ T cells become marginalized by the acquisition of functional defects and reduced cell numbers in a process defined as T cell exhaustion. Similarly, T cell tolerance to self-antigen is also characterized by impaired effector function and eventual deletion of self-reactive T cells. Induction of both tolerance and exhaustion involve many shared inhibitory mechanisms, thus similar therapeutic approaches have proven effective in these distinct environments. We previously demonstrated that tolerant self-reactive CD8+ T cells expressing dual-T cell receptors (i.e., dual-TCR) could be rescued by immunization through a second TCR specific for a foreign antigen. These data revealed that T cell tolerance was regulated at the level of the self-reactive TCR. Here, dual-TCR CD8+ T cells were used to examine if exhaustion during persistent viral infection could be rescued by an analogous strategy of immunization through a second TCR not involved in recognition of virus. In direct contrast to the rescue achievable in tolerant CD8+ T cells, exhausted T cells were equally impaired through both TCR. These findings suggest that exhaustion is maintained by defects downstream of the virus-specific TCR, and establish that exhaustion and tolerance are distinctly regulated states of T cell dysfunction. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
120. Durable Adoptive Immunotherapy for Leukemia Produced by Manipulation of Multiple Regulatory Pathways of CD8+ T-Cell Tolerance.
- Author
-
Berrien-Elliott, Melissa M., Jackson, Stephanie R., Meyer, Jennifer M., Rouskey, Craig J., Nguyen, Thanh-Long M., Yagita, Hideo, Greenberg, Philip D., DiPaolo, Richard J., and Teague, Ryan M.
- Subjects
- *
CD8 antigen , *CANCER treatment , *T cells , *LEUKEMIA treatment , *CANCER immunotherapy , *CANCER invasiveness , *CANCER cell growth - Abstract
Tolerizing mechanisms within the host and tumor microenvironment inhibit T-cell effector functions that can control cancer. These mechanisms blunt adoptive immunotherapy with infused T-cells due to a complex array of signals that determine T-cell tolerance, survival, or deletion. Ligation of the negative regulatory receptors CTLA4, PD-1(PDCD1), or LAG3 on T-cells normally hinders their response to antigen through nonredundant biochemical processes that interfere with stimulatory pathways. In this study, we used an established mouse model of T-cell tolerance to define the roles of these inhibitory receptors in regulating CD8+ T-cell tolerance during adoptive immunotherapy to treat leukemia. Blocking CTLA4 and PD-1 in vivo combined to promote survival of transferred T-cells despite powerful deletional signals that mediate Bim (BCL2L11)-dependent apoptosis. However, this dual blockade was not optimal for stimulating effector function by responding T-cells, which required the additional blockade of LAG3 to induce full expansion and allow the acquisition of robust cytolytic activity. Thus, the cooperation of multiple distinct regulatory pathways was needed for the survival and effector differentiation of adoptively transferred tumor-reactive CD8+ T-cells. Our work defines the immune escape pathways in which simultaneous blockade could yield durable immunotherapeutic responses that can eradicate disseminated leukemia. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
121. Inflammation programs self‐reactive CD8+T cells to acquire T‐box‐mediated effector function but does not prevent deletional tolerance
- Author
-
Jackson, Stephanie R., Yuan, Jinyun, Berrien‐Elliott, Melissa M., Chen, Collin L., Meyer, Jennifer M., Donlin, Maureen J., and Teague, Ryan M.
- Abstract
T‐box transcription factor expression enables effector differentiation of self‐reactive T cells upon antigen engagement, but fails to prevent their deletion. CD8+T cells must detect foreign antigens and differentiate into effector cells to eliminate infections. But, when self‐antigen is recognized instead, mechanisms of peripheral tolerance prevent acquisition of effector function to avoid autoimmunity. These distinct responses are influenced by inflammatory and regulatory clues from the tissue environment, but the mechanism(s) by which naive T cells interpret these signals to generate the appropriate immune response are unclear. The identification of the molecules operative in these cell‐fate decisions is crucial for developing new treatment options for patients with cancer or autoimmunity, where manipulation of T cell activity is desired to alter the course of disease. With the use of an in vivo murine model to examine CD8+T cell responses to healthy self‐tissue, we correlated self‐tolerance with a failure to induce the T‐box transcription factors T‐bet and Eomes. However, inflammation associated with acute microbial infection induced T‐bet and Eomes expression and promoted effector differentiation of self‐reactive T cells under conditions that normally favor tolerance. In the context of a Listeriainfection, these functional responses relied on elevated T‐bet expression, independent of Eomes. Alternatively, infection with LCMV induced higher Eomes expression, which was sufficient in the absence of T‐bet to promote effector cytokine production. Our results place T‐box transcription factors at a molecular crossroads between CD8+T cell anergy and effector function upon recognition of peripheral self‐antigen, and suggest that inflammation during T cell priming directs these distinct cellular responses.
- Published
- 2014
- Full Text
- View/download PDF
122. Flow cytometry-based ex vivomurine NK cell cytotoxicity assay
- Author
-
Wong, Pamela, Wagner, Julia A., Berrien-Elliott, Melissa M., Schappe, Timothy, and Fehniger, Todd A.
- Abstract
Direct killing of diseased cells is a hallmark function of NK cells. This protocol describes a flow-based assay to measure in vivoactivated murine NK cells’ ability to kill target cells ex vivo. Existing published protocols for assaying ex vivoNK cell killing utilized the radioactive chromium release assay or were designed for human NK cells. This protocol details specifically an ex vivocytotoxicity assay using primary murine NK cells enriched from splenocytes that were activated in vivowith poly(I:C).
- Published
- 2021
- Full Text
- View/download PDF
123. Phase I/Dose Expansion Trial of Brentuximab vedotin/Lenalidomide in Relapsed or Refractory Diffuse Large B-cell Lymphoma
- Author
-
Ward, Jeffrey P., Berrien-Elliott, Melissa M., Gomez, Felicia, Luo, Jingqin, Becker-Hapak, Michelle, Cashen, Amanda F., Wagner-Johnston, Nina D., Maddocks, Kami, Mosior, Matthew, Foster, Mark, Krysiak, Kilannin, Schmidt, Alina, Skidmore, Zachary L., Desai, Sweta, Watkins, Marcus P., Fischer, Anne, Griffith, Malachi, Griffith, Obi L., Fehniger, Todd A., and Bartlett, Nancy L.
- Abstract
New therapies are needed for patients with relapsed/refractory (rel/ref) diffuse large B-cell lymphoma (DLBCL) who do not benefit from or are ineligible for stem cell transplant and chimeric antigen receptor therapy. The CD30-targeted antibody-drug conjugate brentuximab vedotin (BV) and the immunomodulator lenalidomide (Len) have demonstrated promising activity as single agents in this population. We report the results of a phase I/dose expansion trial evaluating the combination of BV/Len in rel/ref DLBCL. Thirty-seven patients received BV every 21 days with Len dosed continuously for a maximum of 16 cycles. The maximum tolerated dose of the combination was 1.2 mg/kg BV with 20 mg/day Len. BV/Len was well tolerated with a toxicity profile consistent with their use as single agents. Most patients required G-CSF support due to neutropenia. Overall response rate (ORR) was 57% (95% CI: 39.6-72.5%), complete response rate 35% (95% CI: 20.7-52.6%), median duration of response 13.1 months, median progression-free survival 10.2 (95% CI: 5.5-13.7) months and median overall survival 14.3 months (95% CI 10.2-35.6). Response rates were highest in patients with CD30+ DLBCL (73%) but did not differ according to cell of origin (p=0.96). NK cell expansion and phenotypic changes in CD8+ T-cell subsets in non-responders were identified by mass cytometry. BV/Len represents a potential treatment option for patients with rel/ref DLBCL. This combination is being further explored in a phase III study (NCT04404283). Current trial registered at www.clinicaltrials.gov (NCT02086604).
- Published
- 2021
- Full Text
- View/download PDF
124. Induced CD8a identifies human NK cells with enhanced proliferative fitness and modulates NK cell activation.
- Author
-
Cubitt, Celia C., Wong, Pamela, Dorando, Hannah K., Foltz, Jennifer A., Tran, Jennifer, Marsala, Lynne, Marin, Nancy D., Foster, Mark, Schappe, Timothy, Fatima, Hijab, Becker-Hapak, Michelle, Zhou, Alice Y., Hwang, Kimberly, Jacobs, Miriam T., Russler-Germain, David A., Mace, Emily M., Berrien-Elliott, Melissa M., Payton, Jacqueline E., and Fehniger, Todd A.
- Subjects
- *
KILLER cells , *CELL populations , *CELL physiology - Abstract
The surface receptor CD8α is present on 20%--80% of human (but not mouse) NK cells, yet its function on NK cells remains poorly understood. CD8α expression on donor NK cells was associated with a lack of therapeutic responses in patients with leukemia in prior studies, thus, we hypothesized that CD8α may affect critical NK cell functions. Here, we discovered that CD8α-- NK cells had improved control of leukemia in xenograft models compared with CD8α+ NK cells, likely due to an enhanced capacity for proliferation. Unexpectedly, we found that CD8α expression was induced on approximately 30% of previously CD8α-- NK cells following IL-15 stimulation. These induced CD8α+ (iCD8α+) NK cells had the greatest proliferation, responses to IL-15 signaling, and metabolic activity compared with those that sustained existing CD8α expression (sustained CD8α+) or those that remained CD8α-- (persistent CD8α--). These iCD8α+ cells originated from an IL-15Rβhi NK cell population, with CD8α expression dependent on the transcription factor RUNX3. Moreover, CD8A CRISPR/Cas9 deletion resulted in enhanced responses through the activating receptor NKp30, possibly by modulating KIR inhibitory function. Thus, CD8α status identified human NK cell capacity for IL-15--induced proliferation and metabolism in a time-dependent fashion, and its presence had a suppressive effect on NK cell--activating receptors. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
125. Human CD56brightNK Cells Acquire Potent Anti-Leukemia Functionality Following IL-15 Priming
- Author
-
Wagner, Julia A, Romee, Rizwan, Rosario, Maximillian, Berrien-Elliott, Melissa M, Schneider, Stephanie E, Leong, Jeffrey W, Sullivan, Ryan P, Jewell, Brea A, Schappe, Timothy, Abdel-Latif, Sara, Ireland, Aaron R, Jaishankar, Devika, King, Justin A, Vij, Ravi, and Fehniger, Todd A
- Abstract
Natural killer (NK) cells are innate lymphoid cells that mediate anti-tumor responses via cytotoxicity and effector cytokine production. Human NK cells are divided into two subsets based on relative expression of CD56 (CD56brightand CD56dim) that classically participate in distinct functions. Cytotoxic CD56dimNK cells respond to tumor targets without prior stimulation, resulting in target cell death and transient secretion of effector cytokines (e.g. IFN-γ). In contrast, immunoregulatory CD56brightNK cells secrete abundant IFN-γ and other cytokines in response to cytokine receptor stimulation, but respond minimally to tumor target-based triggering. As a result of this dichotomy, translational strategies to enhance NK cell function for cancer immunotherapy have focused exclusively on the CD56dimsubset.
- Published
- 2016
- Full Text
- View/download PDF
126. First-in-Human Phase 1 study of a CD16A bispecific innate cell engager, AFM24, targeting EGFR-expressing solid tumors.
- Author
-
El-Khoueiry A, Saavedra O, Thomas J, Livings C, Garralda E, Hintzen G, Kohlhas L, Vanosmael D, Koch J, Rajkovic E, Ravenstijn P, Nuciforo P, Fehniger TA, Foster M, Berrien-Elliott MM, Wingert S, Stäble S, Morales-Espinosa D, Rivas D, Emig M, and Lopez J
- Abstract
Purpose: Innate immune cell-based therapies have shown promising antitumor activity against solid and hematologic malignancies. AFM24, a bispecific innate cell engager, binds CD16A on natural killer (NK) cells/macrophages and EGFR on tumor cells, redirecting antitumor activity towards tumors. The safety and tolerability of AFM24 was evaluated in this Phase 1/2a dose escalation/dose expansion study in patients with recurrent or persistent, advanced solid tumors known to express EGFR., Methods: The main objective in Phase 1 was to determine the maximum tolerated dose (MTD) and/or recommended phase 2 dose (RP2D). The primary endpoint was the incidence of dose-limiting toxicities (DLTs) during the observation period. Secondary endpoints included the incidence of treatment emergent adverse events and pharmacokinetics., Results: In the dose escalation phase, 35 patients received AFM24 weekly across seven dose cohorts (14-720 mg). One patient experienced a DLT of Grade 3 infusion-related reaction (IRR). IRRs were mainly reported after the first infusion; these were manageable with premedication and a gradual increase in infusion rate. Pharmacokinetics were dose-proportional and CD16A receptor occupancy on NK cells approached saturation between 320-480 mg. Paired tumor biopsies demonstrated activation of innate and adaptive immune responses within the tumor. Best objective response was stable disease in 10/35 patients; four had stable disease for 4.3-7.1 months., Conclusions: AFM24 was well tolerated with 480 mg established as the RP2D. AFM24 could be a novel therapy for patients with EGFR-expressing solid tumors with suitable tolerability and appropriate pharmacokinetic properties for further development in combination with other immuno-oncology therapeutics.
- Published
- 2025
- Full Text
- View/download PDF
127. A "Prime and Expand" strategy using the multifunctional fusion proteins to generate memory-like NK cells for cell therapy.
- Author
-
Shrestha N, Dee MJ, Chaturvedi P, Leclerc GM, Mathyer M, Dufour C, Arthur L, Becker-Hapak M, Foster M, McClain E, Pena NV, Kage K, Zhu X, George V, Liu B, Egan J, Echeverri C, Wang M, You L, Kong L, Li L, Berrien-Elliott MM, Cooper ML, Fehniger TA, Rhode PR, and Wong HC
- Subjects
- Humans, Animals, Mice, Cell- and Tissue-Based Therapy methods, Immunotherapy, Adoptive methods, Interleukin-15 metabolism, Killer Cells, Natural immunology, Killer Cells, Natural metabolism, Recombinant Fusion Proteins genetics, Immunologic Memory
- Abstract
Adoptive cellular therapy (ACT) using memory-like (ML) natural killer (NK) cells, generated through overnight ex vivo activation with IL-12, IL-15, and IL-18, has shown promise for treating hematologic malignancies. We recently reported that a multifunctional fusion molecule, HCW9201, comprising IL-12, IL-15, and IL-18 domains could replace individual cytokines for priming human ML NK cell programming ("Prime" step). However, this approach does not include ex vivo expansion, thereby limiting the ability to test different doses and schedules. Here, we report the design and generation of a multifunctional fusion molecule, HCW9206, consisting of human IL-7, IL-15, and IL-21 cytokines. We observed > 300-fold expansion for HCW9201-primed human NK cells cultured for 14 days with HCW9206 and HCW9101, an IgG1 antibody, recognizing the scaffold domain of HCW9206 ("Expand" step). This expansion was dependent on both HCW9206 cytokines and interactions of the IgG1 mAb with CD16 receptors on NK cells. The resulting "Prime and Expand" ML NK cells exhibited elevated metabolic capacity, stable epigenetic IFNG promoter demethylation, enhanced antitumor activity in vitro and in vivo, and superior persistence in NSG mice. Thus, the "Prime and Expand" strategy represents a simple feeder cell-free approach to streamline manufacturing of clinical-grade ML NK cells to support multidose and off-the-shelf ACT., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
128. Cytokines drive the formation of memory-like NK cell subsets via epigenetic rewiring and transcriptional regulation.
- Author
-
Foltz JA, Tran J, Wong P, Fan C, Schmidt E, Fisk B, Becker-Hapak M, Russler-Germain DA, Johnson J, Marin ND, Cubitt CC, Pence P, Rueve J, Pureti S, Hwang K, Gao F, Zhou AY, Foster M, Schappe T, Marsala L, Berrien-Elliott MM, Cashen AF, Bednarski JJ, Fertig E, Griffith OL, Griffith M, Wang T, Petti AA, and Fehniger TA
- Subjects
- Humans, Gene Expression Regulation immunology, Cell Differentiation immunology, Interleukin-15 immunology, Killer Cells, Natural immunology, Epigenesis, Genetic immunology, Immunologic Memory immunology, Cytokines immunology
- Abstract
Activation of natural killer (NK) cells with the cytokines interleukin-12 (IL-12), IL-15, and IL-18 induces their differentiation into memory-like (ML) NK cells; however, the underlying epigenetic and transcriptional mechanisms are unclear. By combining ATAC-seq, CITE-seq, and functional analyses, we discovered that IL-12/15/18 activation results in two main human NK fates: reprogramming into enriched memory-like (eML) NK cells or priming into effector conventional NK (effcNK) cells. eML NK cells had distinct transcriptional and epigenetic profiles and enhanced function, whereas effcNK cells resembled cytokine-primed cNK cells. Two transcriptionally discrete subsets of eML NK cells were also identified, eML-1 and eML-2, primarily arising from CD56
bright or CD56dim mature NK cell subsets, respectively. Furthermore, these eML subsets were evident weeks after transfer of IL-12/15/18-activated NK cells into patients with cancer. Our findings demonstrate that NK cell activation with IL-12/15/18 results in previously unappreciated diverse cellular fates and identifies new strategies to enhance NK therapies.- Published
- 2024
- Full Text
- View/download PDF
129. Anti-myeloma efficacy of CAR-iNKT is enhanced with a long-acting IL-7, rhIL-7-hyFc.
- Author
-
O'Neal J, Cooper ML, Ritchey JK, Gladney S, Niswonger J, González LS, Street E, Haas GJ, Carter A, Amayta PN, Gao F, Lee BH, Choi D, Berrien-Elliott M, Zhou A, Fehniger TA, Rettig MP, and DiPersio JF
- Subjects
- Humans, Animals, Mice, Interleukin-7, B-Cell Maturation Antigen, Receptors, Antigen, T-Cell genetics, Multiple Myeloma genetics, Receptors, Chimeric Antigen metabolism, Graft vs Host Disease etiology, Graft vs Host Disease prevention & control
- Abstract
Multiple myeloma (MM), a malignancy of mature plasma cells, remains incurable. B-cell maturation antigen (BCMA) is the lead protein target for chimeric antigen receptor (CAR) therapy because of its high expression in most MM, with limited expression in other cell types, resulting in favorable on-target, off tumor toxicity. The response rate to autologous BCMA CAR-T therapy is high; however, it is not curative and is associated with risks of cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome. Outcomes in patients treated with BCMA CAR-T cells (CAR-Ts) may improve with allogeneic CAR T-cell therapy, which offer higher cell fitness and reduced time to treatment. However, to prevent the risk of graft-versus-host disease (GVHD), allogenic BCMA CAR-Ts require genetic deletion of the T-cell receptor (TCR), which has potential for unexpected functional or phenotype changes. Invariant natural killer T cells (iNKTs) have an invariant TCR that does not cause GVHD and, as a result, can be used in an allogeneic setting without the need for TCR gene editing. We demonstrate significant anti-myeloma activity of BCMA CAR-iNKTs in a xenograft mouse model of myeloma. We found that a long-acting interleukin-7 (IL-7), rhIL-7-hyFc, significantly prolonged survival and reduced tumor burden in BCMA CAR-iNKT-treated mice in both primary and re-challenge settings. Furthermore, in CRS in vitro assays, CAR-iNKTs induced less IL-6 than CAR-Ts, suggesting a reduced likelihood of CAR-iNKT therapy to induce CRS in patients. These data suggest that BCMA CAR-iNKTs are potentially a safer, effective alternative to BCMA CAR-Ts and that BCMA CAR-iNKT efficacy is further potentiated with rhIL-7-hyFc., (© 2023 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
130. Costimulatory domains direct distinct fates of CAR-driven T-cell dysfunction.
- Author
-
Selli ME, Landmann JH, Terekhova M, Lattin J, Heard A, Hsu YS, Chang TC, Chang J, Warrington J, Ha H, Kingston N, Hogg G, Slade M, Berrien-Elliott MM, Foster M, Kersting-Schadek S, Gruszczynska A, DeNardo D, Fehniger TA, Artyomov M, and Singh N
- Subjects
- Humans, Receptors, Antigen, T-Cell genetics, Immunotherapy, Adoptive methods, T-Lymphocytes, Antigens, CD19, Receptors, Chimeric Antigen genetics, Lymphoma etiology
- Abstract
T cells engineered to express chimeric antigen receptors (CARs) targeting CD19 have demonstrated impressive activity against relapsed or refractory B-cell cancers yet fail to induce durable remissions for nearly half of all patients treated. Enhancing the efficacy of this therapy requires detailed understanding of the molecular circuitry that restrains CAR-driven antitumor T-cell function. We developed and validated an in vitro model that drives T-cell dysfunction through chronic CAR activation and interrogated how CAR costimulatory domains, central components of CAR structure and function, contribute to T-cell failure. We found that chronic activation of CD28-based CARs results in activation of classical T-cell exhaustion programs and development of dysfunctional cells that bear the hallmarks of exhaustion. In contrast, 41BB-based CARs activate a divergent molecular program and direct differentiation of T cells into a novel cell state. Interrogation using CAR T cells from a patient with progressive lymphoma confirmed the activation of this novel program in a failing clinical product. Furthermore, we demonstrate that 41BB-dependent activation of the transcription factor FOXO3 is directly responsible for impairing CAR T-cell function. These findings identify that costimulatory domains are critical regulators of CAR-driven T-cell failure and that targeted interventions are required to overcome costimulation-dependent dysfunctional programs., (© 2023 by The American Society of Hematology.)
- Published
- 2023
- Full Text
- View/download PDF
131. Immunotherapeutic approach to reduce senescent cells and alleviate senescence-associated secretory phenotype in mice.
- Author
-
Shrestha N, Chaturvedi P, Zhu X, Dee MJ, George V, Janney C, Egan JO, Liu B, Foster M, Marsala L, Wong P, Cubitt CC, Foltz JA, Tran J, Schappe T, Hsiao K, Leclerc GM, You L, Echeverri C, Spanoudis C, Carvalho A, Kanakaraj L, Gilkes C, Encalada N, Kong L, Wang M, Fang B, Wang Z, Jiao JA, Muniz GJ, Jeng EK, Valdivieso N, Li L, Deth R, Berrien-Elliott MM, Fehniger TA, Rhode PR, and Wong HC
- Subjects
- Mice, Animals, Aging, Inflammation, Immunotherapy, Phenotype, Senescence-Associated Secretory Phenotype, Cellular Senescence genetics
- Abstract
Accumulation of senescent cells (SNCs) with a senescence-associated secretory phenotype (SASP) has been implicated as a major source of chronic sterile inflammation leading to many age-related pathologies. Herein, we provide evidence that a bifunctional immunotherapeutic, HCW9218, with capabilities of neutralizing TGF-β and stimulating immune cells, can be safely administered systemically to reduce SNCs and alleviate SASP in mice. In the diabetic db/db mouse model, subcutaneous administration of HCW9218 reduced senescent islet β cells and SASP resulting in improved glucose tolerance, insulin resistance, and aging index. In naturally aged mice, subcutaneous administration of HCW9218 durably reduced the level of SNCs and SASP, leading to lower expression of pro-inflammatory genes in peripheral organs. HCW9218 treatment also reverted the pattern of key regulatory circadian gene expression in aged mice to levels observed in young mice and impacted genes associated with metabolism and fibrosis in the liver. Single-nucleus RNA Sequencing analysis further revealed that HCW9218 treatment differentially changed the transcriptomic landscape of hepatocyte subtypes involving metabolic, signaling, cell-cycle, and senescence-associated pathways in naturally aged mice. Long-term survival studies also showed that HCW9218 treatment improved physical performance without compromising the health span of naturally aged mice. Thus, HCW9218 represents a novel immunotherapeutic approach and a clinically promising new class of senotherapeutic agents targeting cellular senescence-associated diseases., (© 2023 HCW Biologics Inc, Washington University School of Medicine and Nova Southeastern University. Aging Cell published by Anatomical Society and John Wiley & Sons Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
132. Allogeneic natural killer cell therapy.
- Author
-
Berrien-Elliott MM, Jacobs MT, and Fehniger TA
- Subjects
- Humans, Immunotherapy, Adoptive methods, Cytokine Release Syndrome, Killer Cells, Natural, Neoplasms therapy, Hematopoietic Stem Cell Transplantation
- Abstract
Interest in adoptive cell therapy for treating cancer is exploding owing to early clinical successes of autologous chimeric antigen receptor (CAR) T lymphocyte therapy. However, limitations using T cells and autologous cell products are apparent as they (1) take weeks to generate, (2) utilize a 1:1 donor-to-patient model, (3) are expensive, and (4) are prone to heterogeneity and manufacturing failures. CAR T cells are also associated with significant toxicities, including cytokine release syndrome, immune effector cell-associated neurotoxicity syndrome, and prolonged cytopenias. To overcome these issues, natural killer (NK) cells are being explored as an alternative cell source for allogeneic cell therapies. NK cells have an inherent ability to recognize cancers, mediate immune functions of killing and communication, and do not induce graft-versus-host disease, cytokine release syndrome, or immune effector cell-associated neurotoxicity syndrome. NK cells can be obtained from blood or cord blood or be derived from hematopoietic stem and progenitor cells or induced pluripotent stem cells, and can be expanded and cryopreserved for off-the-shelf availability. The first wave of point-of-care NK cell therapies led to the current allogeneic NK cell products being investigated in clinical trials with promising preliminary results. Basic advances in NK cell biology and cellular engineering have led to new translational strategies to block inhibition, enhance and broaden target cell recognition, optimize functional persistence, and provide stealth from patients' immunity. This review details NK cell biology, as well as NK cell product manufacturing, engineering, and combination therapies explored in the clinic leading to the next generation of potent, off-the-shelf cellular therapies for blood cancers., (© 2023 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
133. Combining AFM13, a Bispecific CD30/CD16 Antibody, with Cytokine-Activated Blood and Cord Blood-Derived NK Cells Facilitates CAR-like Responses Against CD30 + Malignancies.
- Author
-
Kerbauy LN, Marin ND, Kaplan M, Banerjee PP, Berrien-Elliott MM, Becker-Hapak M, Basar R, Foster M, Garcia Melo L, Neal CC, McClain E, Daher M, Nunez Cortes AK, Desai S, Inng Lim FW, Mendt MC, Schappe T, Li L, Shaim H, Shanley M, Ensley EL, Uprety N, Wong P, Liu E, Ang SO, Cai R, Nandivada V, Mohanty V, Miao Q, Shen Y, Baran N, Fowlkes NW, Chen K, Muniz-Feliciano L, Champlin RE, Nieto YL, Koch J, Treder M, Fischer W, Okamoto OK, Shpall EJ, Fehniger TA, and Rezvani K
- Subjects
- Humans, Blood drug effects, Blood immunology, Cells, Cultured, Combined Modality Therapy, Cytokines pharmacology, Fetal Blood drug effects, Fetal Blood immunology, Ki-1 Antigen immunology, Receptors, IgG immunology, Antibodies, Bispecific therapeutic use, Immunotherapy methods, Killer Cells, Natural immunology, Leukemia therapy, Lymphoma therapy
- Abstract
Purpose: Natural killer (NK)-cell recognition and function against NK-resistant cancers remain substantial barriers to the broad application of NK-cell immunotherapy. Potential solutions include bispecific engagers that target NK-cell activity via an NK-activating receptor when simultaneously targeting a tumor-specific antigen, as well as enhancing functionality using IL12/15/18 cytokine pre-activation., Experimental Design: We assessed single-cell NK-cell responses stimulated by the tetravalent bispecific antibody AFM13 that binds CD30 on leukemia/lymphoma targets and CD16A on various types of NK cells using mass cytometry and cytotoxicity assays. The combination of AFM13 and IL12/15/18 pre-activation of blood and cord blood-derived NK cells was investigated in vitro and in vivo ., Results: We found heterogeneity within AFM13-directed conventional blood NK cell (cNK) responses, as well as consistent AFM13-directed polyfunctional activation of mature NK cells across donors. NK-cell source also impacted the AFM13 response, with cNK cells from healthy donors exhibiting superior responses to those from patients with Hodgkin lymphoma. IL12/15/18-induced memory-like NK cells from peripheral blood exhibited enhanced killing of CD30
+ lymphoma targets directed by AFM13, compared with cNK cells. Cord-blood NK cells preactivated with IL12/15/18 and ex vivo expanded with K562-based feeders also exhibited enhanced killing with AFM13 stimulation via upregulation of signaling pathways related to NK-cell effector function. AFM13-NK complex cells exhibited enhanced responses to CD30+ lymphomas in vitro and in vivo ., Conclusions: We identify AFM13 as a promising combination with cytokine-activated adult blood or cord-blood NK cells to treat CD30+ hematologic malignancies, warranting clinical trials with these novel combinations., (©2021 American Association for Cancer Research.)- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.