26 results on '"Sweta Desai"'
Search Results
2. Supplementary Table 4 from A Fusion Protein Complex that Combines IL-12, IL-15, and IL-18 Signaling to Induce Memory-Like NK Cells for Cancer Immunotherapy
- Author
-
Todd A. Fehniger, Hing C. Wong, Melissa M. Berrien-Elliott, Ryan P. Sullivan, Caitlin A. Prendes, Emily K. Jeng, Gabriela J. Muniz, Jin-an Jiao, Peter R. Rhode, Jack O. Egan, Lijing You, Laritza L. Ramirez, Christian A. Echeverri, Victor L. Gallo, Catherine M. Spanoudis, Xiaoyun Zhu, David A. Russler-Germain, Julia A. Wagner, Pamela Wong, Patrick Pence, Carly C. Neal, Sweta Desai, Jennifer Tran, Timothy Schappe, Mark Foster, Lynne I. Marsala, Gilles M. Leclerc, Pallavi Chaturvedi, Michael J. Dee, Ethan McClain, Niraj Shrestha, and Michelle K. Becker-Hapak
- Abstract
Table 4 Excel Document
- Published
- 2023
3. Supplementary Figures 1-8 and Tables 1-3 from A Fusion Protein Complex that Combines IL-12, IL-15, and IL-18 Signaling to Induce Memory-Like NK Cells for Cancer Immunotherapy
- Author
-
Todd A. Fehniger, Hing C. Wong, Melissa M. Berrien-Elliott, Ryan P. Sullivan, Caitlin A. Prendes, Emily K. Jeng, Gabriela J. Muniz, Jin-an Jiao, Peter R. Rhode, Jack O. Egan, Lijing You, Laritza L. Ramirez, Christian A. Echeverri, Victor L. Gallo, Catherine M. Spanoudis, Xiaoyun Zhu, David A. Russler-Germain, Julia A. Wagner, Pamela Wong, Patrick Pence, Carly C. Neal, Sweta Desai, Jennifer Tran, Timothy Schappe, Mark Foster, Lynne I. Marsala, Gilles M. Leclerc, Pallavi Chaturvedi, Michael J. Dee, Ethan McClain, Niraj Shrestha, and Michelle K. Becker-Hapak
- Abstract
Supplemental Figures 1-8, Supplemental Tables 1-3
- Published
- 2023
4. Data from A Fusion Protein Complex that Combines IL-12, IL-15, and IL-18 Signaling to Induce Memory-Like NK Cells for Cancer Immunotherapy
- Author
-
Todd A. Fehniger, Hing C. Wong, Melissa M. Berrien-Elliott, Ryan P. Sullivan, Caitlin A. Prendes, Emily K. Jeng, Gabriela J. Muniz, Jin-an Jiao, Peter R. Rhode, Jack O. Egan, Lijing You, Laritza L. Ramirez, Christian A. Echeverri, Victor L. Gallo, Catherine M. Spanoudis, Xiaoyun Zhu, David A. Russler-Germain, Julia A. Wagner, Pamela Wong, Patrick Pence, Carly C. Neal, Sweta Desai, Jennifer Tran, Timothy Schappe, Mark Foster, Lynne I. Marsala, Gilles M. Leclerc, Pallavi Chaturvedi, Michael J. Dee, Ethan McClain, Niraj Shrestha, and Michelle K. Becker-Hapak
- Abstract
Natural killer (NK) cells are a promising cellular therapy for cancer, with challenges in the field including persistence, functional activity, and tumor recognition. Briefly, priming blood NK cells with recombinant human (rh)IL-12, rhIL-15, and rhIL-18 (12/15/18) results in memory-like NK cell differentiation and enhanced responses against cancer. However, the lack of available, scalable Good Manufacturing Process (GMP)–grade reagents required to advance this approach beyond early-phase clinical trials is limiting. To address this challenge, we developed a novel platform centered upon an inert tissue factor scaffold for production of heteromeric fusion protein complexes (HFPC). The first use of this platform combined IL-12, IL-15, and IL-18 receptor engagement (HCW9201), and the second adds CD16 engagement (HCW9207). This unique HFPC expression platform was scalable with equivalent protein quality characteristics in small- and GMP-scale production. HCW9201 and HCW9207 stimulated activation and proliferation signals in NK cells, but HCW9207 had decreased IL-18 receptor signaling. RNA sequencing and multidimensional mass cytometry revealed parallels between HCW9201 and 12/15/18. HCW9201 stimulation improved NK cell metabolic fitness and resulted in the DNA methylation remodeling characteristic of memory-like differentiation. HCW9201 and 12/15/18 primed similar increases in short-term and memory-like NK cell cytotoxicity and IFNγ production against leukemia targets, as well as equivalent control of leukemia in NSG mice. Thus, HFPCs represent a protein engineering approach that solves many problems associated with multisignal receptor engagement on immune cells, and HCW9201-primed NK cells can be advanced as an ideal approach for clinical GMP-grade memory-like NK cell production for cancer therapy.
- Published
- 2023
5. Supplementary Figure Legends from A Fusion Protein Complex that Combines IL-12, IL-15, and IL-18 Signaling to Induce Memory-Like NK Cells for Cancer Immunotherapy
- Author
-
Todd A. Fehniger, Hing C. Wong, Melissa M. Berrien-Elliott, Ryan P. Sullivan, Caitlin A. Prendes, Emily K. Jeng, Gabriela J. Muniz, Jin-an Jiao, Peter R. Rhode, Jack O. Egan, Lijing You, Laritza L. Ramirez, Christian A. Echeverri, Victor L. Gallo, Catherine M. Spanoudis, Xiaoyun Zhu, David A. Russler-Germain, Julia A. Wagner, Pamela Wong, Patrick Pence, Carly C. Neal, Sweta Desai, Jennifer Tran, Timothy Schappe, Mark Foster, Lynne I. Marsala, Gilles M. Leclerc, Pallavi Chaturvedi, Michael J. Dee, Ethan McClain, Niraj Shrestha, and Michelle K. Becker-Hapak
- Abstract
Legends for Supplementary Figures 1-8 and Supplementary Tables 1-5
- Published
- 2023
6. Supplementary Table 5 from A Fusion Protein Complex that Combines IL-12, IL-15, and IL-18 Signaling to Induce Memory-Like NK Cells for Cancer Immunotherapy
- Author
-
Todd A. Fehniger, Hing C. Wong, Melissa M. Berrien-Elliott, Ryan P. Sullivan, Caitlin A. Prendes, Emily K. Jeng, Gabriela J. Muniz, Jin-an Jiao, Peter R. Rhode, Jack O. Egan, Lijing You, Laritza L. Ramirez, Christian A. Echeverri, Victor L. Gallo, Catherine M. Spanoudis, Xiaoyun Zhu, David A. Russler-Germain, Julia A. Wagner, Pamela Wong, Patrick Pence, Carly C. Neal, Sweta Desai, Jennifer Tran, Timothy Schappe, Mark Foster, Lynne I. Marsala, Gilles M. Leclerc, Pallavi Chaturvedi, Michael J. Dee, Ethan McClain, Niraj Shrestha, and Michelle K. Becker-Hapak
- Abstract
Table 5 Excel Document
- Published
- 2023
7. Supplementary Figures from Multidimensional Analyses of Donor Memory-Like NK Cells Reveal New Associations with Response after Adoptive Immunotherapy for Leukemia
- Author
-
Todd A. Fehniger, John DiPersio, Mark A. Schroeder, Keith E. Stockerl-Goldstein, Iskra Pusic, Meagan A. Jacoby, Peter Westervelt, Geoffrey L. Uy, Camille N. Abboud, Rizwan Romee, Feng Gao, Sridhar Nonavinkere Srivatsan, Natalia Jaeger, Matthew L. Cooper, Jennifer A. Foltz, Michelle Becker-Hapak, Ethan McClain, Sweta Desai, Timothy Schappe, Mark Foster, Julia A. Wagner, Pamela Wong, Carly C. Neal, Celia C. Cubitt, Amanda F. Cashen, and Melissa M. Berrien-Elliott
- Abstract
Supplemental figures S1-S11
- Published
- 2023
8. Supplementary Methods from Multidimensional Analyses of Donor Memory-Like NK Cells Reveal New Associations with Response after Adoptive Immunotherapy for Leukemia
- Author
-
Todd A. Fehniger, John DiPersio, Mark A. Schroeder, Keith E. Stockerl-Goldstein, Iskra Pusic, Meagan A. Jacoby, Peter Westervelt, Geoffrey L. Uy, Camille N. Abboud, Rizwan Romee, Feng Gao, Sridhar Nonavinkere Srivatsan, Natalia Jaeger, Matthew L. Cooper, Jennifer A. Foltz, Michelle Becker-Hapak, Ethan McClain, Sweta Desai, Timothy Schappe, Mark Foster, Julia A. Wagner, Pamela Wong, Carly C. Neal, Celia C. Cubitt, Amanda F. Cashen, and Melissa M. Berrien-Elliott
- Abstract
Supplementary patient data, supplementary materials and methods
- Published
- 2023
9. Supplementary Tables from Multidimensional Analyses of Donor Memory-Like NK Cells Reveal New Associations with Response after Adoptive Immunotherapy for Leukemia
- Author
-
Todd A. Fehniger, John DiPersio, Mark A. Schroeder, Keith E. Stockerl-Goldstein, Iskra Pusic, Meagan A. Jacoby, Peter Westervelt, Geoffrey L. Uy, Camille N. Abboud, Rizwan Romee, Feng Gao, Sridhar Nonavinkere Srivatsan, Natalia Jaeger, Matthew L. Cooper, Jennifer A. Foltz, Michelle Becker-Hapak, Ethan McClain, Sweta Desai, Timothy Schappe, Mark Foster, Julia A. Wagner, Pamela Wong, Carly C. Neal, Celia C. Cubitt, Amanda F. Cashen, and Melissa M. Berrien-Elliott
- Abstract
Supplemental patient tables (1-3) and mass cytometry reagent table (4)
- Published
- 2023
10. Synthesis of Pteridine Derivatives as Anti-TB Agents
- Author
-
Sweta Desai, Zarna Dedania, and Naishadh Solanki
- Subjects
Pteridine ,Mycobacterium tuberculosis ,Anti-Tb - Abstract
Tuberculosis (TB) is a major global health issue with a high mortality rate, particularly due to the emergence of drug-resistant strains of Mycobacterium tuberculosis. Hence, the development of novel anti-TB agents is of great significance. In this regard, pteridine derivatives have emerged as a promising scaffold for the development of novel drugs. This study focuses on the synthesis of N-phenyl-6,7-dip-tolylpteridin-4-amine derivatives, and their evaluation for anti-TB activity. The three-step synthesis process involved the preparation of 6,7-dip-tolylpteridin-4-ol from 4,5-diamino-6-hydroxypyrimidine and 4,4-dimethylbenzil, followed by its reaction with phosphoryl chloride and dimethylformamide to yield 4-chloro-6,7-dip-tolylpteridine, and finally, the reaction of the latter with aniline in dimethylformamide to obtain N-phenyl-6,7-dip-tolylpteridin-4-amine derivatives. The synthesized compounds were evaluated for their anti-TB activity against standard drugs streptomycin, ciprofloxacin, and pyrazinamide. The results showed that all the compounds demonstrated significant anti-TB activity, with some displaying activity comparable to the reference drugs. Specifically, compounds E6, E8, E13, and E17 showed activity like that of streptomycin and ciprofloxacin, while compounds E3, E25, E9, E14, E19, and E4 demonstrated activity similar to pyrazinamide. In conclusion, the study highlights the potential of pteridine derivatives as promising anti-TB agents. The synthesized compounds exhibited significant anti-TB activity and could serve as an alternative to current standard therapies., 1.Gagneux S. Ecology and evolution of Mycobacterium tuberculosis. Nature Reviews Microbiology. 2018;16(4):202-13. 2. Parija SC. Genus Mycobacterium and Mycobacterium tuberculosis. Textbook of Microbiology and Immunology: Springer; 2023. p. 419-37. 3. van der Werf TS, de Reus YA. Personalized Tuberculosis Care for Drug-Resistant Tuberculosis. Tuberculosis: Integrated Studies for a Complex Disease: Springer; 2023. p. 403-28. 4. Mitku AA, Dessie ZG, Muluneh EK, Workie DL. Prevalence and associated factors of TB/HIV co-infection among HIV Infected patients in Amhara International Journal of Pharmacy and Biological Sciences Sweta Desai* et al www.ijpbs.com or www.ijpbsonline.com 73 ISSN: 2230-7605 (Online); ISSN: 2321-3272 (Print) Int J Pharm Biol Sci. region, Ethiopia. African health sciences. 2016;16(2):588-95. 5. Saha I, Paul B. Private sector involvement envisaged in the National Strategic Plan for Tuberculosis Elimination 2017–2025: Can Tuberculosis Health Action Learning Initiative model act as a road map? medical journal armed forces india. 2019;75(1):25-7. 6. Verbeeck RK, Günther G, Kibuule D, Hunter C, Rennie TW. Optimizing treatment outcome of first-line anti-tuberculosis drugs: the role of therapeutic drug monitoring. European journal of clinical pharmacology. 2016; 72:905-16. 7. Saravanan M, Niguse S, Abdulkader M, Tsegay E, Hailekiros H, Gebrekidan A, et al. Review on emergence of drug-resistant tuberculosis (MDR & XDR-TB) and its molecular diagnosis in Ethiopia. Microbial pathogenesis. 2018; 117:237-42. 8. Roca I, Akova M, Baquero F, Carlet J, Cavaleri M, Coenen S, et al. The global threat of antimicrobial resistance: science for intervention. New microbes and new infections. 2015; 6:22-9. 9. Brigden G, Hewison C, Varaine F. New developments in the treatment of drug-resistant tuberculosis: clinical utility of bedaquiline and delamanid. Infection and drug resistance. 2015:367-78. 10. Nguyen T, Cao T, Akkerman O, Tiberi S, Vu D, Alffenaar J. Bedaquiline as part of combination therapy in adults with pulmonary multi-drug resistant tuberculosis. Expert review of clinical pharmacology. 2016;9(8):1025-37. 11. Ferlazzo G, Mohr E, Laxmeshwar C, Hewison C, Hughes J, Jonckheere S, et al. Early safety and efficacy of the combination of bedaquiline and delamanid for the treatment of patients with drug-resistant tuberculosis in Armenia, India, and South Africa: a retrospective cohort study. The Lancet Infectious Diseases. 2018;18(5):536-44. 12. Cox V, McKenna L, Acquah R, Reuter A, Wasserman S, Vambe D, et al. Clinical perspectives on treatment of rifampicin-resistant/multidrug-resistant TB. The International Journal of Tuberculosis and Lung Disease. 2020;24(11):1134-44. 13. Vasava MS, Bhoi MN, Rathwa SK, Borad MA, Nair SG, Patel HD. Drug development against tuberculosis: past, present and future. Indian journal of tuberculosis. 2017;64(4):252-75. 14. Stubbs B, Siddiqi K, Elsey H, Siddiqi N, Ma R, Romano E, et al. Tuberculosis and non-communicable disease multimorbidity: an analysis of the world health survey in 48 low-and middle-income countries. International Journal of Environmental Research and Public Health. 2021;18(5):2439. 15. De Coen LM, Heugebaert TS, Garcia D, Stevens CV. Synthetic entries to and biological activity of pyrrolopyrimidines. Chemical Reviews. 2016;116(1):80-139. 16. Wróbel A, Drozdowska D. Recent design and structure-activity relationship studies on the modifications of DHFR inhibitors as anticancer agents. Current Medicinal Chemistry. 2021;28(5):910-39. 17. Xiao Z, Morris‐Natschke SL, Lee KH. Strategies for the optimization of natural leads to anticancer drugs or drug candidates. Medicinal research reviews. 2016;36(1):32-91. 18. Fernández-Villa D, Aguilar MR, Rojo L. Folic acid antagonists: antimicrobial and immunomodulating mechanisms and applications. International journal of molecular sciences. 2019;20(20):4996. 19. Sumaily KM, Mujamammi AH. Phenylketonuria: A new look at an old topic, advances in laboratory diagnosis, and therapeutic strategies. International journal of health sciences. 2017;11(5):63. 20. Carmona‐Martínez V, Ruiz‐Alcaraz AJ, Vera M, Guirado A, Martínez‐Esparza M, García‐Peñarrubia P. Therapeutic potential of pteridine derivatives: A comprehensive review. Medicinal Research Reviews. 2019;39(2):461-516. 21. Reddy GS, Pal M. Indole derivatives as anti-tubercular agents: An overview on their synthesis and biological activities. Current Medicinal Chemistry. 2021;28(22):4531-68.
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Table S1 from 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
-
Katayoun Rezvani, Todd A. Fehniger, Elizabeth J. Shpall, Oswaldo Keith Okamoto, Wolfgang Fischer, Martin Treder, Joachim Koch, Yago L. Nieto, Richard E. Champlin, Luis Muniz-Feliciano, Ken Chen, Natalie W. Fowlkes, Natalia Baran, Yifei Shen, Qi Miao, Vakul Mohanty, Vandana Nandivada, Rong Cai, Sonny O. Ang, Enli Liu, Pamela Wong, Nadima Uprety, Emily L. Ensley, Mayra Shanley, Hila Shaim, Li Li, Timothy Schappe, Mayela Carolina Mendt, Francesca Wei Inng Lim, Sweta Desai, Ana Karen Nunez Cortes, May Daher, Ethan McClain, Carly C. Neal, Luciana Garcia Melo, Mark Foster, Rafet Basar, Michelle Becker-Hapak, Melissa M. Berrien-Elliott, Pinaki P. Banerjee, Mecit Kaplan, Nancy D. Marin, and Lucila N. Kerbauy
- Abstract
Supplementary Table S1
- Published
- 2023
12. Supplementary Figure S3 from 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
-
Katayoun Rezvani, Todd A. Fehniger, Elizabeth J. Shpall, Oswaldo Keith Okamoto, Wolfgang Fischer, Martin Treder, Joachim Koch, Yago L. Nieto, Richard E. Champlin, Luis Muniz-Feliciano, Ken Chen, Natalie W. Fowlkes, Natalia Baran, Yifei Shen, Qi Miao, Vakul Mohanty, Vandana Nandivada, Rong Cai, Sonny O. Ang, Enli Liu, Pamela Wong, Nadima Uprety, Emily L. Ensley, Mayra Shanley, Hila Shaim, Li Li, Timothy Schappe, Mayela Carolina Mendt, Francesca Wei Inng Lim, Sweta Desai, Ana Karen Nunez Cortes, May Daher, Ethan McClain, Carly C. Neal, Luciana Garcia Melo, Mark Foster, Rafet Basar, Michelle Becker-Hapak, Melissa M. Berrien-Elliott, Pinaki P. Banerjee, Mecit Kaplan, Nancy D. Marin, and Lucila N. Kerbauy
- Abstract
Supplementary Fig. S3. Differentiation of ML NK cells and Preactivated-Expanded CB-NK cells.
- Published
- 2023
13. Supplementary Materials and Methods from 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
-
Katayoun Rezvani, Todd A. Fehniger, Elizabeth J. Shpall, Oswaldo Keith Okamoto, Wolfgang Fischer, Martin Treder, Joachim Koch, Yago L. Nieto, Richard E. Champlin, Luis Muniz-Feliciano, Ken Chen, Natalie W. Fowlkes, Natalia Baran, Yifei Shen, Qi Miao, Vakul Mohanty, Vandana Nandivada, Rong Cai, Sonny O. Ang, Enli Liu, Pamela Wong, Nadima Uprety, Emily L. Ensley, Mayra Shanley, Hila Shaim, Li Li, Timothy Schappe, Mayela Carolina Mendt, Francesca Wei Inng Lim, Sweta Desai, Ana Karen Nunez Cortes, May Daher, Ethan McClain, Carly C. Neal, Luciana Garcia Melo, Mark Foster, Rafet Basar, Michelle Becker-Hapak, Melissa M. Berrien-Elliott, Pinaki P. Banerjee, Mecit Kaplan, Nancy D. Marin, and Lucila N. Kerbauy
- Abstract
Supplementary Materials and Methods
- Published
- 2023
14. Data from 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
-
Katayoun Rezvani, Todd A. Fehniger, Elizabeth J. Shpall, Oswaldo Keith Okamoto, Wolfgang Fischer, Martin Treder, Joachim Koch, Yago L. Nieto, Richard E. Champlin, Luis Muniz-Feliciano, Ken Chen, Natalie W. Fowlkes, Natalia Baran, Yifei Shen, Qi Miao, Vakul Mohanty, Vandana Nandivada, Rong Cai, Sonny O. Ang, Enli Liu, Pamela Wong, Nadima Uprety, Emily L. Ensley, Mayra Shanley, Hila Shaim, Li Li, Timothy Schappe, Mayela Carolina Mendt, Francesca Wei Inng Lim, Sweta Desai, Ana Karen Nunez Cortes, May Daher, Ethan McClain, Carly C. Neal, Luciana Garcia Melo, Mark Foster, Rafet Basar, Michelle Becker-Hapak, Melissa M. Berrien-Elliott, Pinaki P. Banerjee, Mecit Kaplan, Nancy D. Marin, and Lucila N. Kerbauy
- 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.
- Published
- 2023
15. Phase 1/dose expansion trial of brentuximab vedotin and lenalidomide in relapsed or refractory diffuse large B-cell lymphoma
- Author
-
Nancy L. Bartlett, Felicia Gomez, Matthew Mosior, Malachi Griffith, Anne Fischer, Michelle Becker-Hapak, Mark P. Foster, Melissa M. Berrien-Elliott, Alina D. Schmidt, Marcus Watkins, Amanda F. Cashen, Kilannin Krysiak, Kami J. Maddocks, Nina D. Wagner-Johnston, Jingqin Luo, Obi L. Griffith, Zachary L. Skidmore, Todd A. Fehniger, Sweta Desai, and Jeffrey P. Ward
- Subjects
medicine.medical_specialty ,Immunoconjugates ,CD30 ,Clinical Trials and Observations ,Immunology ,Population ,Neutropenia ,Biochemistry ,Gastroenterology ,Refractory ,Internal medicine ,medicine ,Humans ,Refractory Diffuse Large B-Cell Lymphoma ,Brentuximab vedotin ,education ,Lenalidomide ,Brentuximab Vedotin ,education.field_of_study ,business.industry ,Cell Biology ,Hematology ,medicine.disease ,Lymphoma ,Treatment Outcome ,Lymphoma, Large B-Cell, Diffuse ,Neoplasm Recurrence, Local ,business ,medicine.drug - 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 1/dose expansion trial evaluating the combination of BV/Len in rel/ref DLBCL. Thirty-seven patients received BV every 21 days, with Len administered continuously for a maximum of 16 cycles. The maximum tolerated dose of the combination was 1.2 mg/kg BV with 20 mg/d Len. BV/Len was well tolerated with a toxicity profile consistent with their use as single agents. Most patients required granulocyte colony-stimulating factor support because of neutropenia. The overall response rate 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 months (95% CI, 5.5-13.7); and median overall survival, 14.3 months (95% CI, 10.2-35.6). Response rates were highest in patients with CD30+ DLBCL (73%), but they did not differ according to cell of origin (P = .96). NK cell expansion and phenotypic changes in CD8+ T-cell subsets in nonresponders 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 3 study (registered on https://clinicaltrials.org as NCT04404283). This trial was registered on https://clinicaltrials.gov as NCT02086604.
- Published
- 2022
16. Systemic IL-15 promotes allogeneic cell rejection in patients treated with natural killer cell adoptive therapy
- Author
-
Ethan McClain, Sweta Desai, Amanda F. Cashen, Mark P. Foster, Peter Westervelt, Patrick Soon-Shiong, Michelle Becker-Hapak, Keith Stockerl-Goldstein, Jeffrey S. Miller, Miriam T. Jacobs, Mark A. Schroeder, Pamela Wong, Camille N. Abboud, Patrick Pence, Geoffrey L. Uy, Feng Gao, Claudio G. Brunstein, Melissa M. Berrien-Elliott, Meagan A. Jacoby, Iskra Pusic, Sarah Cooley, John F. DiPersio, and Todd A. Fehniger
- Subjects
Male ,Adoptive cell transfer ,Recombinant Fusion Proteins ,T cell ,Lymphocyte ,Immunology ,Antineoplastic Agents ,CD8-Positive T-Lymphocytes ,Immunotherapy, Adoptive ,Biochemistry ,Natural killer cell ,Cell therapy ,Humans ,Medicine ,Cytotoxic T cell ,Interleukin-15 ,business.industry ,Allogeneic Cells ,Hematopoietic Stem Cell Transplantation ,Cell Biology ,Hematology ,Killer Cells, Natural ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Interleukin 15 ,Cancer research ,Female ,business ,CD8 - Abstract
Natural killer (NK) cells are a promising alternative to T cells for cancer immunotherapy. Adoptive therapies with allogeneic, cytokine-activated NK cells are being investigated in clinical trials. However, the optimal cytokine support after adoptive transfer to promote NK cell expansion, and persistence remains unclear. Correlative studies from 2 independent clinical trial cohorts treated with major histocompatibility complex-haploidentical NK cell therapy for relapsed/refractory acute myeloid leukemia revealed that cytokine support by systemic interleukin-15 (IL-15; N-803) resulted in reduced clinical activity, compared with IL-2. We hypothesized that the mechanism responsible was IL-15/N-803 promoting recipient CD8 T-cell activation that in turn accelerated donor NK cell rejection. This idea was supported by increased proliferating CD8+ T-cell numbers in patients treated with IL-15/N-803, compared with IL-2. Moreover, mixed lymphocyte reactions showed that IL-15/N-803 enhanced responder CD8 T-cell activation and proliferation, compared with IL-2 alone. Additionally, IL-15/N-803 accelerated the ability of responding T cells to kill stimulator-derived memory-like NK cells, demonstrating that additional IL-15 can hasten donor NK cell elimination. Thus, systemic IL-15 used to support allogeneic cell therapy may paradoxically limit their therapeutic window of opportunity and clinical activity. This study indicates that stimulating patient CD8 T-cell allo-rejection responses may critically limit allogeneic cellular therapy supported with IL-15. This trial was registered at www.clinicaltrials.gov as #NCT03050216 and #NCT01898793.
- Published
- 2022
17. A Fusion Protein Complex that Combines IL-12, IL-15, and IL-18 Signaling to Induce Memory-Like NK Cells for Cancer Immunotherapy
- Author
-
Pamela Wong, Jin-An Jiao, Timothy Schappe, Gilles M. Leclerc, Sweta Desai, Gabriela J. Muniz, Melissa M. Berrien-Elliott, Mark P. Foster, Lynne Marsala, Pallavi Chaturvedi, Carly Neal, Niraj Shrestha, Christian A. Echeverri, Xiaoyun Zhu, David A. Russler-Germain, Caitlin A. Prendes, Catherine M. Spanoudis, Todd A. Fehniger, Ethan McClain, Jack O. Egan, Michael J. Dee, Peter R. Rhode, Hing C. Wong, Jennifer Tran, Laritza L. Ramirez, Ryan P. Sullivan, Lijing You, Victor L. Gallo, Julia A. Wagner, Emily K. Jeng, Michelle Becker-Hapak, and Patrick Pence
- Subjects
Cancer Research ,Recombinant Fusion Proteins ,medicine.medical_treatment ,Immunology ,Cell ,CD16 ,Article ,Cell therapy ,Mice ,Immune system ,Cancer immunotherapy ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Interleukin-15 ,Leukemia ,Chemistry ,Remission Induction ,Interleukin-18 ,Interleukin-12 ,Xenograft Model Antitumor Assays ,Fusion protein ,Cell biology ,Killer Cells, Natural ,medicine.anatomical_structure ,Interleukin 15 ,Interleukin 12 ,Receptors, Natural Killer Cell ,Immunologic Memory - Abstract
Natural killer (NK) cells are a promising cellular therapy for cancer, with challenges in the field including persistence, functional activity, and tumor recognition. Briefly, priming blood NK cells with recombinant human (rh)IL-12, rhIL-15, and rhIL-18 (12/15/18) results in memory-like NK cell differentiation and enhanced responses against cancer. However, the lack of available, scalable Good Manufacturing Process (GMP)–grade reagents required to advance this approach beyond early-phase clinical trials is limiting. To address this challenge, we developed a novel platform centered upon an inert tissue factor scaffold for production of heteromeric fusion protein complexes (HFPC). The first use of this platform combined IL-12, IL-15, and IL-18 receptor engagement (HCW9201), and the second adds CD16 engagement (HCW9207). This unique HFPC expression platform was scalable with equivalent protein quality characteristics in small- and GMP-scale production. HCW9201 and HCW9207 stimulated activation and proliferation signals in NK cells, but HCW9207 had decreased IL-18 receptor signaling. RNA sequencing and multidimensional mass cytometry revealed parallels between HCW9201 and 12/15/18. HCW9201 stimulation improved NK cell metabolic fitness and resulted in the DNA methylation remodeling characteristic of memory-like differentiation. HCW9201 and 12/15/18 primed similar increases in short-term and memory-like NK cell cytotoxicity and IFNγ production against leukemia targets, as well as equivalent control of leukemia in NSG mice. Thus, HFPCs represent a protein engineering approach that solves many problems associated with multisignal receptor engagement on immune cells, and HCW9201-primed NK cells can be advanced as an ideal approach for clinical GMP-grade memory-like NK cell production for cancer therapy.
- Published
- 2021
18. Multidimensional Analyses of Donor Memory-Like NK Cells Reveal New Associations with Response after Adoptive Immunotherapy for Leukemia
- Author
-
Amanda F. Cashen, Matthew L. Cooper, Carly Neal, Camille N. Abboud, Feng Gao, Sweta Desai, Melissa M. Berrien-Elliott, Michelle Becker-Hapak, Celia C. Cubitt, Timothy Schappe, Pamela Wong, Mark A. Schroeder, John F. DiPersio, Peter Westervelt, Julia A. Wagner, Jennifer A. Foltz, Meagan A. Jacoby, Natalia Jaeger, Iskra Pusic, Mark P. Foster, Geoffrey L. Uy, Todd A. Fehniger, Ethan McClain, Rizwan Romee, Keith Stockerl-Goldstein, and Sridhar Nonavinkere Srivatsan
- Subjects
0301 basic medicine ,business.industry ,medicine.medical_treatment ,Myeloid leukemia ,Cancer ,Immunotherapy ,medicine.disease ,Immunotherapy, Adoptive ,Phenotype ,Article ,Killer Cells, Natural ,Cell therapy ,Leukemia, Myeloid, Acute ,03 medical and health sciences ,Leukemia ,030104 developmental biology ,0302 clinical medicine ,Oncology ,In vivo ,030220 oncology & carcinogenesis ,Cancer research ,Humans ,Medicine ,Mass cytometry ,business - Abstract
Natural killer (NK) cells are an emerging cancer cellular therapy and potent mediators of antitumor immunity. Cytokine-induced memory-like (ML) NK cellular therapy is safe and induces remissions in patients with acute myeloid leukemia (AML). However, the dynamic changes in phenotype that occur after NK-cell transfer that affect patient outcomes remain unclear. Here, we report comprehensive multidimensional correlates from ML NK cell–treated patients with AML using mass cytometry. These data identify a unique in vivo differentiated ML NK–cell phenotype distinct from conventional NK cells. Moreover, the inhibitory receptor NKG2A is a dominant, transcriptionally induced checkpoint important for ML, but not conventional NK-cell responses to cancer. The frequency of CD8α+ donor NK cells is negatively associated with AML patient outcomes after ML NK therapy. Thus, elucidating the multidimensional dynamics of donor ML NK cells in vivo revealed critical factors important for clinical response, and new avenues to enhance NK-cell therapeutics. Significance: Mass cytometry reveals an in vivo memory-like NK-cell phenotype, where NKG2A is a dominant checkpoint, and CD8α is associated with treatment failure after ML NK–cell therapy. These findings identify multiple avenues for optimizing ML NK–cell immunotherapy for cancer and define mechanisms important for ML NK–cell function. This article is highlighted in the In This Issue feature, p. 1775
- Published
- 2020
19. Hematopoietic cell transplantation donor-derived memory-like NK cells functionally persist after transfer into patients with leukemia
- Author
-
Melissa M. Berrien-Elliott, Jennifer A. Foltz, David A. Russler-Germain, Carly C. Neal, Jennifer Tran, Margery Gang, Pamela Wong, Bryan Fisk, Celia C. Cubitt, Nancy D. Marin, Alice Y. Zhou, Miriam T. Jacobs, Mark Foster, Timothy Schappe, Ethan McClain, Samantha Kersting-Schadek, Sweta Desai, Patrick Pence, Michelle Becker-Hapak, Jeremy Eisele, Matthew Mosior, Lynne Marsala, Obi L. Griffith, Malachi Griffith, Saad M. Khan, David H. Spencer, John F. DiPersio, Rizwan Romee, Geoffrey L. Uy, Camille N. Abboud, Armin Ghobadi, Peter Westervelt, Keith Stockerl-Goldstein, Mark A. Schroeder, Fei Wan, Wen-Rong Lie, Patrick Soon-Shiong, Allegra A. Petti, Amanda F. Cashen, and Todd A. Fehniger
- Subjects
Interleukin-15 ,Killer Cells, Natural ,Leukemia, Myeloid, Acute ,Hematopoietic Stem Cell Transplantation ,Humans ,General Medicine ,Article ,Immunity, Innate - Abstract
Natural killer (NK) cells are innate lymphoid cells that eliminate cancer cells, produce cytokines, and are being investigated as a nascent cellular immunotherapy. Impaired NK cell function, expansion, and persistence remain key challenges for optimal clinical translation. One promising strategy to overcome these challenges is cytokine-induced memory-like (ML) differentiation, whereby NK cells acquire enhanced antitumor function after stimulation with interleukin-12 (IL-12), IL-15, and IL-18. Here, reduced-intensity conditioning (RIC) for HLA -haploidentical hematopoietic cell transplantation (HCT) was augmented with same-donor ML NK cells on day +7 and 3 weeks of N-803 (IL-15 superagonist) to treat patients with relapsed/refractory acute myeloid leukemia (AML) in a clinical trial (NCT02782546). In 15 patients, donor ML NK cells were well tolerated, and 87% of patients achieved a composite complete response at day +28, which corresponded with clearing high-risk mutations, including TP53 variants. NK cells were the major blood lymphocytes for 2 months after HCT with 1104-fold expansion (over 1 to 2 weeks). Phenotypic and transcriptional analyses identified donor ML NK cells as distinct from conventional NK cells and showed that ML NK cells persisted for over 2 months. ML NK cells expressed CD16, CD57, and high granzyme B and perforin, along with a unique transcription factor profile. ML NK cells differentiated in patients had enhanced ex vivo function compared to conventional NK cells from both patients and healthy donors. Overall, same-donor ML NK cell therapy with 3 weeks of N-803 support safely augmented RIC haplo-HCT for AML.
- Published
- 2022
20. Neoantigen Landscape Supports Feasibility of Personalized Cancer Vaccine for Follicular Lymphoma
- Author
-
Cody A. Ramirez, Felix Frenkel, Michelle Becker-Hapak, Erica K. Barnell, Ethan D. McClain, Sweta Desai, Timothy Schappe, Onyinyechi C. Onyeador, Olga Kudryashova, Vladislav Belousov, Alexander Bagaev, Elena Ocheredko, Susanna Kiwala, Jasreet Hundal, Zachary L. Skidmore, Marcus P. Watkins, Thomas B. Mooney, Jason R. Walker, Kilannin Krysiak, David A. Russler-Germain, Felicia Gomez, Catrina C. Fronick, Robert S. Fulton, Robert D. Schreiber, Neha Mehta-Shah, Amanda F. Cashen, Brad S. Kahl, Ravshan Ataullakhanov, Nancy L. Bartlett, Malachi Griffith, Obi L. Griffith, and Todd A. Fehniger
- Abstract
Personalized cancer vaccines designed to target neoantigens represent a promising new treatment paradigm in oncology. In contrast to classical idiotype vaccines, we hypothesized that ‘polyvalent’ vaccines could be engineered for the personalized treatment of follicular lymphoma (FL) using neoantigen discovery by combined whole exome sequencing (WES) and RNA sequencing (RNA-Seq). Fifty-eight tumor samples from 57 patients with FL underwent WES and RNA-Seq. Somatic and B-cell clonotype neoantigens were predicted and filtered to identify high-quality neoantigens. B-cell clonality was determined by alignment of B-cell receptor (BCR) CDR3 regions from RNA-Seq data, grouping at the protein level, and comparison to the BCR repertoire of RNA-Seq data from healthy individuals. An average of 52 somatic mutations per patient (range: 2-172) were identified, and two or more (median: 15) high-quality neoantigens were predicted for 56 of 58 samples. The predicted neoantigen peptides were composed of missense mutations (76%), indels (9%), gene fusions (3%), and BCR sequences (11%). Building off of these preclinical analyses, we initiated a pilot clinical trial using personalized neoantigen vaccination combined with PD-1 blockade in patients with relapsed or refractory FL (#NCT03121677). Synthetic long peptide (SLP) vaccines were successfully synthesized for and administered to all four patients enrolled to date. Initial results demonstrate feasibility, safety, and potential immunologic and clinical responses. Our study suggests that a genomics-driven personalized cancer vaccine strategy is feasible for patients with FL, and this may overcome prior challenges in the field.
- Published
- 2022
21. Donor memory-like NK cells persist and induce remissions in pediatric patients with relapsed AML after transplant
- Author
-
Jeffrey J. Bednarski, Clare Zimmerman, Melissa M. Berrien-Elliott, Jennifer A. Foltz, Michelle Becker-Hapak, Carly C. Neal, Mark Foster, Timothy Schappe, Ethan McClain, Patrick P. Pence, Sweta Desai, Samantha Kersting-Schadek, Pamela Wong, David A. Russler-Germain, Bryan Fisk, Wen-Rong Lie, Jeremy Eisele, Stephanie Hyde, Sima T. Bhatt, Obi L. Griffith, Malachi Griffith, Allegra A. Petti, Amanda F. Cashen, and Todd A. Fehniger
- Subjects
Killer Cells, Natural ,Leukemia, Myeloid, Acute ,Immunology ,Hematopoietic Stem Cell Transplantation ,Humans ,Transplantation, Homologous ,Cell Biology ,Hematology ,Child ,Unrelated Donors ,Biochemistry - Abstract
Pediatric and young adult (YA) patients with acute myeloid leukemia (AML) who relapse after allogeneic hematopoietic cell transplantation (HCT) have an extremely poor prognosis. Standard salvage chemotherapy and donor lymphocyte infusions (DLIs) have little curative potential. Previous studies showed that natural killer (NK) cells can be stimulated ex vivo with interleukin-12 (IL-12), -15, and -18 to generate memory-like (ML) NK cells with enhanced antileukemia responses. We treated 9 pediatric/YA patients with post-HCT relapsed AML with donor ML NK cells in a phase 1 trial. Patients received fludarabine, cytarabine, and filgrastim followed 2 weeks later by infusion of donor lymphocytes and ML NK cells from the original HCT donor. ML NK cells were successfully generated from haploidentical and matched-related and -unrelated donors. After infusion, donor-derived ML NK cells expanded and maintained an ML multidimensional mass cytometry phenotype for >3 months. Furthermore, ML NK cells exhibited persistent functional responses as evidenced by leukemia-triggered interferon-γ production. After DLI and ML NK cell adoptive transfer, 4 of 8 evaluable patients achieved complete remission at day 28. Two patients maintained a durable remission for >3 months, with 1 patient in remission for >2 years. No significant toxicity was experienced. This study demonstrates that, in a compatible post-HCT immune environment, donor ML NK cells robustly expand and persist with potent antileukemic activity in the absence of exogenous cytokines. ML NK cells in combination with DLI present a novel immunotherapy platform for AML that has relapsed after allogeneic HCT. This trial was registered at https://clinicaltrials.gov as #NCT03068819.
- Published
- 2021
22. 531 AFM13-targeted blood and cord-blood-derived memory-like NK cells as therapy for CD30+ malignancies
- Author
-
Mark P. Foster, Rong Cai, Ana Karen Nunez Cortes, Yago Nieto, Vakul Mohanty, Melissa M. Berrien-Elliott, Sweta Desai, Joachim Koch, Michelle Becker-Hapak, Mecit Kaplan, Pamela Wong, May Daher, Natalie W. Fowlkes, Martin Treder, Luciana Garcia Melo, Wolfgang Fischer, Vandana Nandivada, Carly Neal, Luis Muniz-Feliciano, Li Li, Sonny Ang, Natalia Baran, Enli Liu, Elizabeth J. Shpall, Todd A. Fehniger, Yifei Shen, Richard E. Champlin, Ethan McClain, Mayela Mendt, Pinaki P. Banerjee, Francesca Lorraine Wei Inng Lim, Mayra Hernandez Sanabria, Oswaldo Keith Okamoto, Hila Shaim, Lucila Nassif Kerbauy, Katy Rezvani, Nancy D. Marin, Rafet Basar, Timothy Schappe, and Ken Chen
- Subjects
CD30 ,Cell ,Biology ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,lcsh:RC254-282 ,Lymphoma ,Cell therapy ,Leukemia ,medicine.anatomical_structure ,Antigen ,Cord blood ,medicine ,Cancer research ,NSG mouse - Abstract
Background Natural killer (NK) cells are a nascent cellular immunotherapy for hematologic malignancies. Target recognition of NK cell-resistant cancers remains a substantial barrier to broad application of NK cell therapy. One solution are bispecific engagers that trigger NK cells via an NK activating receptor when simultaneously engaging a tumor-specific antigen. Methods Here, we investigated single NK cell responses stimulated by the tetravalent bispecific innate cell engager (ICE®) AFM13 that binds CD30 on leukemia/lymphoma targets and CD16A on several types of NK cells. Results Multidimensional mass cytometry revealed heterogeneity within AFM13-directed conventional (c)NK cell responses, as well as consistent polyfunctional activation of mature terminally differentiated NK cells across donors. The source of NK cells also impacted the AFM13 response, with cNK cells from healthy donors exhibiting superior responses to those from Hodgkin lymphoma patients. IL-12, IL-15, and IL-18-induced memory-like NK cells from peripheral blood exhibited enhanced killing of CD30+ lymphoma targets directed by AFM13, compared to cNK cells. Cord-blood expanded NK cells that were pre-activated with IL-12, IL-15 and IL-18 also exhibited enhanced killing with AFM13 stimulation, via upregulation of signaling pathways related to NK cell effector function. These cells were stably pre-loaded with AFM13 enhancing responses to CD30+ lymphomas in vitro and in vivo in immunodeficient NSG mouse models. Conclusions Collectively, these data identify promising combinations of AFM13 with cytokine-activated adult blood or cord blood NK cells against CD30+ hematologic malignancies, warranting clinical trials with these novel combinations.
- Published
- 2020
23. Cytokine-Induced Memory-like NK Cells Have a Distinct Single Cell Transcriptional Profile and Persist for Months in Adult and Pediatric Leukemia Patients after Adoptive Transfer
- Author
-
Bryan Fisk, Pamela Wong, Jennifer A. Foltz, Margery Gang, Obi L. Griffith, Matthew Mosior, Nancy D. Marin, Jeffrey J. Bednarski, Lynne Marsala, Melissa M. Berrien-Elliott, Timothy Schappe, Amanda F. Cashen, Mark P. Foster, Alice Zhou, Miriam T. Jacobs, Clare Zimmerman, David A. Russler-Germain, Malachi Griffith, Celia C. Cubitt, Sweta Desai, Jennifer Tran, Michelle Becker-Hapak, Todd A. Fehniger, Ethan McClain, Allegra A. Petti, Patrick Pence, Saad M. Khan, and Carly Neal
- Subjects
Pediatric leukemia ,Adoptive cell transfer ,business.industry ,medicine.medical_treatment ,Immunology ,Cell ,Cell Biology ,Hematology ,Biochemistry ,medicine.anatomical_structure ,Cytokine ,Medicine ,business - Abstract
Natural killer (NK) cells are innate lymphoid cells that mediate anti-tumor responses and exhibit innate memory following stimulation with IL-12, IL-15, and IL-18, thereby differentiating into cytokine-induced memory-like (ML) NK cells. ML NK cells have well-described enhanced anti-tumor properties; however, the molecular mechanisms underlying their enhanced functionality are not well-understood. Initial reports of allogeneic donor ML NK cellular therapy for relapsed/refractory (rel/ref) acute myeloid leukemia (AML) demonstrated safety and a 47% CR/CRi rate (PMID32826231). In this setting, allogeneic ML NK cells are rejected after 3 weeks by recipient T cells, which precludes long-term evaluation of their biology. To address this limitation, we conducted a clinical trial for rel/ref AML patients that added adoptive transfer of same-donor ML NK cells on day +7 of a reduced-intensity conditioning (RIC) MHC-haploidentical HCT, followed by 4 doses of IL-15 (N-803) over 2 weeks (NCT02782546). Since the ML NK cells are from the HCT donor, they are not rejected, but remain MHC-haploidentical to the patient leukemia. Using samples from these patients, we profiled the single cell transcriptomes of NK cells using multidimensional CITE-seq, combining scRNAseq with a custom NK panel of antibodies. To identify donor ML NK cells in an unbiased fashion, we developed a CITE-seq ML NK classifier from in vitro differentiated paired conventional NK (cNK) and ML NK cells. This classifier was applied via transfer learning to CITE-seq analyzed samples from the donor (cNK cells) and patients at days +28 and +60. This approach identified 28-40% of NK cells as ML at Day +28 post-HCT. Only 1-6% of donor peripheral blood NK cells and 4-7% of NK cells in comparator leukemia patients at day +28 after conventional haplo-HCT alone were identified as ML NK cells (Fig 1A). These ML NK cells had a cell surface receptor profile analogous to a previously reported mass cytometry phenotype. Within the CITE-seq data, ML NK cells expressed a transcriptional profile consistent with enhanced functionality (GZMK, GZMA, GNLY), secreted proteins (LTB, CKLF), a distinct adhesome, and evidence of prior activation (MHC Class II and interferon-inducible genes). ML NK cells had a unique NK receptor repertoire including increased KIR2DL4, KLRC1(NKG2A), CD300A, NCAM1(CD56) , and CD2 with decreased expression of the inhibitory receptor KLRB1(CD161). Furthermore, ML NK cells upregulated HOPX, a transcription factor implicated in memory T cells and murine CMV adaptive NK cells. Additionally, ML NK cells downregulated transcription factors related to terminal maturation (ZEB2) and exhaustion (NR4A2). We next sought to identify changes during ML differentiation in patients post-HCT from day +28 to +60 post-HCT. Trajectory analysis identified a ML NK cell state distinct from cNK cells that was present at least 60 days post-HCT (Fig 1B). The ML transcriptional phenotype continued to modulate during late differentiation, including downregulation of GZMK and NCAM1, and upregulation of maturation related transcription factors, while maintaining high expression of HOPX. ML NK cells retained their enhanced functionality during in vivo differentiation, as patient ML NK cells had significantly increased IFNγ production compared to cNK cells after restimulation with leukemia targets or cytokines using mass cytometry (Fig. 2). Subsequently, we confirmed the ML CITE-seq profile in an independent clinical trial treating pediatric AML relapsed after allogenic HCT with same-donor ML NK cells (NCT03068819). In this setting, ML NK cells expressed a similar transcriptional signature and persisted for at least 2 months in the absence of exogenous cytokine support. Thus, ML NK cells possess a distinct transcriptional and surface proteomic profile and undergo in vivo differentiation while persisting within patients for at least 2 months. These findings reveal novel and unique aspects of the ML NK cell molecular program, as well as their prolonged functional persistence in vivo in patients, assisting in future clinical trial design. Figure 1 Figure 1. Disclosures Foltz: Kiadis: Patents & Royalties: TGFbeta expanded NK cells; EMD Millipore: Other: canine antibody licensing fees. Berrien-Elliott: Wugen: Consultancy, Patents & Royalties: 017001-PRO1, Research Funding. Bednarski: Horizon Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees. Fehniger: Wugen: Consultancy, Current equity holder in publicly-traded company, Patents & Royalties: related to memory like NK cells, Research Funding; ImmunityBio: Research Funding; Kiadis: Other; Affimed: Research Funding; Compass Therapeutics: Research Funding; HCW Biologics: Research Funding; OrcaBio: Other; Indapta: Other.
- Published
- 2021
24. Assessment of quality of life in parents of epileptic children and its associated factors
- Author
-
Rameshkumar Dihora, Sweta Desai, and Ashish Goti
- Subjects
Gerontology ,Quality of life (healthcare) ,business.industry ,Medicine ,business - Abstract
Background: Aim of the study was to assess quality of life (QOL) of parents of epileptic child and its association with various factors like demographic, clinical, therapeutic and behavioral factors.Methods: Consenting parents of 160 epileptic children were enrolled after finishing child’s visit to the pediatrician. Parents were enquired on baseline demographic variables like age, gender, socio-economic status, parental education; clinical details like type of epilepsy, duration of seizure, seizure frequency and co-morbidity and therapeutic factors like treatment of epilepsy and adverse drug reactions. QOL was evaluated using QOLCE questionnaire and Childhood Illness-related Parenting Stress Inventory and analysed.Results: Out of 160 parents, 85% belong to 30-40 years of age and 62.5% were male. Deteriorated quality of life was reported by parents as mean score 63.46±7.69. QOL was significantly poor (p
- Published
- 2020
25. Cytokine-Induced Memory-like (ML) NK Cells Persist for > 2 Months Following Adoptive Transfer into Leukemia Patients with a MHC-Compatible Hematopoietic Cell Transplant (HCT)
- Author
-
Amanda F. Cashen, Sweta Desai, Mark P. Foster, Tim Schappe, Michelle Becker-Hapak, Carly Neal, Todd A. Fehniger, Ethan McClain, Melissa M. Berrien-Elliott, and Jennifer A. Foltz
- Subjects
education.field_of_study ,Adoptive cell transfer ,biology ,business.industry ,Immunology ,Population ,Cell Biology ,Hematology ,Biochemistry ,Perforin ,Granzyme ,Interleukin 15 ,Interleukin 12 ,biology.protein ,Medicine ,Tumor necrosis factor alpha ,Granzyme M ,education ,business - Abstract
Natural killer (NK) cells exhibit innate memory or memory-like responses following stimulation with haptens, viruses, or cytokines. Human memory-like (ML) NK cells differentiate following a short-term activation with IL-12, IL-15, and IL-18, and have increased anti-tumor activity against AML and other cancers in vitro, in xenograft models, and in the first-in human phase 1 clinical trial of ML NK cells in AML (PMID27655849). In this trial, CR/Cri was observed in >50% of patients treated, and mass cytometry revealed a unique multi-dimensional phenotype of in vivo differentiated ML NK cells that was confirmed using donor-specific HLA markers. Although adoptively transferred MHC-haploidentical ML NK cells expanded and differentiated over 2-3 weeks, these cells were eliminated by recipient allogeneic immune responses, a challenge observed with all allogeneic lymphocyte therapies. The immune rejection observed in the allogeneic setting precluded following ML NK phenotype, persistence, and function long-term in these patients. We hypothesized that ML NK cells could persist longer than 2-3 weeks in an MHC-compatible setting, and thus be able to assess ML NK cells durability. To test this idea, a phase 2 clinical trial was designed for relapsed/refractory AML patients, who receive a reduced-intensity HLA-haploidentical hematopoietic cell transplant (HCT), followed by same-donor ML NK cell adoptive transfer at day 7, with 2 weeks of IL-15/N-803 support (NCT02782546). Using mass cytometry, ML NK cells were confirmed as distinct from conventional NK cells, CD56hi/NKp30hi/CD62Lhi/KIR+/NKG2A+/CD57+/-, by viSNE analysis, and clearly inconsistent with immature NK cells arising from the HCT graft (CD56bright/KIR-/CD57-). ML NK cells persisted in patients for at least 2 months (n=5) following adoptive transfer, and constituted 20-50% of total NK cells at day 60 (n=3, 206±97 cells/μl; mean±SEM; peak ML NK cells = 751-1106 cells/µl, D21-D28, n=5). These ML NK cells appeared highly functional (56±8% IFN-γ+, 20±3% TNF+, 41±7% CD107a+) when stimulated with tumor targets immediately ex vivo on study day 28 (n=7). Unsupervised clustering of scRNA-seq from patient samples acquired 14-60 days after ML NK cell adoptive transfer identified a subset of NK cells transcriptionally distinct from conventional CD56bright and CD56dim. This NK cell population was the majority of NK cells at study Day 21 and remained identifiable 2 months post-transfer. In agreement with the mass cytometry data, these NK cells expressed high levels of KIRs with scRNA-seq analysis uncovering novel transcriptional changes in granzyme M, perforin, KLRG1, and IFNG suggesting ML NK cells represent a mature, activated NK cell subset distinct from conventional NK cells arising from the graft. In addition, scRNAseq analysis identified high expression of the transcription factor RUNX3, a potential regulator of ML NK cell phenotype in vivo. In conclusion, a single infusion of ML NK cells resulted in a durable population of highly functional NK cells, as evidenced by multi-dimensional analyses using mass cytometry and scRNA-sequencing. These studies provide evidence that ML NK cell therapy in the MHC-compatible setting overcomes persistence barriers and provide a platform for innovation in NK cell therapeutics. Disclosures Cashen: Celgene: Other: Speaker's Bureau; Seattle Genetics: Other: Speaker's Bureau; Novartis: Other: Speaker's Bureau. Fehniger:Cyto-Sen Therapeutics: Consultancy; Horizon Pharma PLC: Other: Consultancy (Spouse).
- Published
- 2019
26. Adoptively Transferred Donor-Derived Cytokine Induced Memory-like NK Cells Persist and Induce Remission in Pediatric Patient with Relapsed Acute Myeloid Leukemia after Hematopoietic Cell Transplantation
- Author
-
Sweta Desai, Clare Zimmerman, Melissa M. Berrien-Elliott, Jeffrey J. Bednarski, Tim Schappe, Todd A. Fehniger, Amanda F. Cashen, Ethan McClain, Michelle Becker-Hapak, and Mark P. Foster
- Subjects
Adoptive cell transfer ,business.industry ,medicine.medical_treatment ,Immunology ,Myeloid leukemia ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,medicine.disease ,Biochemistry ,Granulocyte colony-stimulating factor ,Transplantation ,Leukemia ,hemic and lymphatic diseases ,medicine ,Interleukin 12 ,Cytarabine ,business ,medicine.drug - Abstract
Acute myeloid leukemia (AML) accounts for 18% of pediatric leukemias. For high-risk AML, standard treatment includes multi-agent chemotherapy followed allogeneic hematopoietic cell transplantation (HCT). Despite a high remission rate, 50% of patients relapse and have a poor prognosis with < 20% of patients surviving more than 3 years. Salvage chemotherapy alone or combined with donor lymphocyte infusions (DLI) have little curative potential, and new treatment strategies are needed for relapsed-refractory AML. Previous studies have shown that natural killer (NK) cells can be stimulated ex vivo with IL-12/15/18 to generate a memory-like phenotype with enhanced anti-leukemia effect. In adults with relapsed-refractory AML, adoptive transfer of MHC-haploidentical cytokine-induced memory-like (CIML or ML) NK cells induced remission in 54% of patients (PMID27655849). The infused donor ML NK cells expand in vivo but are rapidly eliminated following recovery of recipient T cells, providing a window of therapeutic activity of 2-3 weeks. We sought to test the safety and efficacy of ML NK cells for treatment of pediatric/young adult patients with post-HCT relapsed AML. We hypothesized that ML NK cells derived from the HCT donor would be well-tolerated, exhibit anti-leukemia activity, and expand with prolonged persistence following transfer into pediatric AML patients. Here, we report the results of the first pediatric patient treated on a phase I clinical trial using ML NK cell therapy for relapsed AML after allogeneic HCT (NCT03068819). Briefly, patients are treated with FLAG (fludarabine, cytarabine and granulocyte colony stimulating factor) salvage chemotherapy to reduce the bulk of AML and provide lymphodepletion for ML NK cell expansion. Two weeks after chemotherapy, a non-mobilized leukapheresis product is collected from the original HCT donor and processed into a T cell-based DLI and ML NK cells. The T cell DLI (1 x 106 T cells/kg) is immediately infused, and the ML NK cells are generated by stimulation with IL-12/15/18 ex vivo for 12-16 hours and then infused (10x106/kg). An 18-month-old male with infant AML had relapse of his leukemia 3 months after MHC-haploidentical HCT. Treatment with chemotherapy, including mitoxantrone and daunorubicin-cytarabine liposome, failed to induce remission. At the time of enrollment on the phase I trial, he had AML blasts in his bone marrow (Table 1). He was treated with FLAG chemotherapy followed by infusion of DLI and ML NK cells from the original haploidentical HCT donor. Assessment at 30 days, 3 months and 6 months post NK cell infusion showed complete remission with no evidence of leukemia and full donor engraftment. Remarkably, donor-derived ML NK cells expanded to 77% of donor lymphocytes on day 28 and still comprised 24% percent of lymphocytes at 6 months post infusion (Figure 1A-C). The expanded donor NK cell phenotype was consistent with ML NK cells (e.g., NKG2A+KIR+) utilizing CyTOF multidimensional analysis previously confirmed to identify ML NK cells (Figure 1D). The ML NK cells were functional as demonstrated by leukemia-triggered IFN-γ production immediately ex vivo from day 7-28 samples (Figure 1E-F). The patient's clinical course was complicated by mild gastrointestinal graft-versus-host disease that resolved with low-dose steroids and tociluzimab. These early results demonstrate proof-of-principle that adoptive transfer of donor-derived ML NK cells in combination with DLI is feasible and offers a novel immunotherapy option for patients with relapsed AML after HCT. Moreover, in this T and NK cell compatible immune environment post-HCT, donor ML NK cells expand and persist robustly in vivo for > 6 months without exogenous cytokine support and have potent anti-leukemic activity. Thus, ML NK cells are a cellular therapy platform to treat AML that has relapsed after allogeneic HCT. Disclosures Cashen: Celgene: Other: Speaker's Bureau; Seattle Genetics: Other: Speaker's Bureau; Novartis: Other: Speaker's Bureau. Fehniger:Horizon Pharma PLC: Other: Consultancy (Spouse); Cyto-Sen Therapeutics: Consultancy.
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.