1
|
Srinagesh H, Jackson C, Shiraz P, Jeyakumar N, Hamilton M, Egeler E, Mavroukakis S, Kuo A, Cancilla J, Sahaf B, Agarwal N, Kanegai A, Kramer AM, Arai S, Bharadwaj S, Dahiya S, Hosoya H, Johnston L, Kennedy V, Liedtke M, Lowsky R, Mikkilineni L, Negrin R, Rezvani A, Sidana S, Shizuru J, Smith M, Weng WK, Feldman S, Frank MJ, Lee Z, Tagliaferri M, Marcondes AM, Miklos D, Mackall C, Muffly L. A phase 1 clinical trial of NKTR-255 with CD19-22 CAR T-cell therapy for refractory B-cell acute lymphoblastic leukemia. Blood 2024; 144:1689-1698. [PMID: 38968138 PMCID: PMC11522888 DOI: 10.1182/blood.2024024952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 07/07/2024] Open
Abstract
ABSTRACT Although chimeric antigen receptor (CAR) T-cell (CAR-T) therapy has revolutionized the treatment of B-cell malignancies, many patients relapse and therefore strategies to improve antitumor immunity are needed. We previously designed a novel autologous bispecific CAR targeting CD19 and CD22 (CAR19-22), which was well tolerated and associated with high response rates but relapse was common. Interleukin-15 (IL15) induces proliferation of diverse immune cells and can augment lymphocyte trafficking. Here, we report the results of a phase 1 clinical trial of the first combination of a novel recombinant polymer-conjugated IL15 receptor agonist (NKTR-255), with CAR19-22, in adults with relapsed/refractory B-cell acute lymphoblastic leukemia. Eleven patients were enrolled, 9 of whom successfully received CAR19-22 followed by NKTR-255. There were no dose-limiting toxicities, with transient fever and myelosuppression as the most common possibly related toxicities. We observed favorable efficacy with 8 of 9 patients (89%) achieving measurable residual disease-negative remission. At 12 months, progression-free survival for NKTR-255 was double that of historical controls (67% vs 38%). We performed correlative analyses to investigate the effects of IL15 receptor agonism. Cytokine profiling showed significant increases in IL15 and the chemokines CXCL9 and CXCL10. The increase in chemokines was associated with decreases in absolute lymphocyte counts and CD8+ CAR T cells in the blood and 10-fold increases in cerebrospinal fluid CAR-T cells, suggesting lymphocyte trafficking to tissue. Combining NKTR-255 with CAR19-22 was safe, feasible, and associated with high rates of durable responses. This trial was registered at www.clinicaltrials.gov as #NCT03233854.
Collapse
Affiliation(s)
| | - Clayton Jackson
- Division of Hematology/Oncology, UT Southwestern, Dallas, TX
| | - Parveen Shiraz
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | | | - Mark Hamilton
- Division of Hematology, Stanford University, Stanford, CA
| | - Emily Egeler
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | | | - Adam Kuo
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | | | - Bita Sahaf
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | - Neha Agarwal
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Alyssa Kanegai
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Anne Marijn Kramer
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | - Sally Arai
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Sushma Bharadwaj
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Saurabh Dahiya
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Hitomi Hosoya
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Laura Johnston
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Vanessa Kennedy
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | | | - Robert Lowsky
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Lekha Mikkilineni
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Robert Negrin
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Andrew Rezvani
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Surbhi Sidana
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Judith Shizuru
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Melody Smith
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Wen-Kai Weng
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Steven Feldman
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | - Matthew J. Frank
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | | | | | | | - David Miklos
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | - Crystal Mackall
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | - Lori Muffly
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| |
Collapse
|
2
|
Pozzi S, Satchi-Fainaro R. The role of CCL2/CCR2 axis in cancer and inflammation: The next frontier in nanomedicine. Adv Drug Deliv Rev 2024; 209:115318. [PMID: 38643840 DOI: 10.1016/j.addr.2024.115318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 04/23/2024]
Abstract
The communication between cells and their microenvironment represents an intrinsic and essential attribute that takes place in several biological processes, including tissue homeostasis and tissue repair. Among these interactions, inflammation is certainly a central biological response that occurs through cytokines and the crosstalk with their respective receptors. In particular, the interaction between CCL2 and its main receptor, CCR2, plays a pivotal role in both harmful and protective inflammatory states, including cancer-mediated inflammation. The activation of the CCL2/CCR2 axis was shown to dictate the migration of macrophages with immune-suppressive phenotype and to aggravate the progression of different cancer types. In addition, this interaction mediates metastasis formation, further limiting the potential therapeutic outcome of anti-cancer drugs. Attempts to inhibit pharmacologically the CCL2/CCR2 axis have yet to show its anti-cancer efficacy as a single agent, but it sheds light on its role as a powerful tool to selectively alleviate pro-tumorigenic and anti-repair inflammation. In this review, we will elucidate the role of CCL2/CCR2 axis in promoting cancer inflammation by activating the host pro-tumorigenic phenotype. Moreover, we will provide some insight into the potential therapeutic benefit of targeting the CCL2/CCR2 axis for cancer and inflammation using novel delivery systems, aiming to sensitize non-responders to currently approved immunotherapies and offer new combinatory approaches.
Collapse
Affiliation(s)
- Sabina Pozzi
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; Sagol School of Neurosciences, Tel Aviv University, Tel Aviv 6997801, Israel.
| |
Collapse
|
3
|
Körtge A, Breitrück A, Doß S, Hofrichter J, Nelz SC, Krüsemann H, Wasserkort R, Fitzner B, Hecker M, Mitzner S, Zettl UK. The Utility of Miniaturized Adsorbers in Exploring the Cellular and Molecular Effects of Blood Purification: A Pilot Study with a Focus on Immunoadsorption in Multiple Sclerosis. Int J Mol Sci 2024; 25:2590. [PMID: 38473836 DOI: 10.3390/ijms25052590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/02/2024] [Accepted: 02/09/2024] [Indexed: 03/14/2024] Open
Abstract
Immunoadsorption (IA) has proven to be clinically effective in the treatment of steroid-refractory multiple sclerosis (MS) relapses, but its mechanism of action remains unclear. We used miniaturized adsorber devices with a tryptophan-immobilized polyvinyl alcohol (PVA) gel sorbent to mimic the IA treatment of patients with MS in vitro. The plasma was screened before and after adsorption with regard to disease-specific mediators, and the effect of the IA treatment on the migration of neutrophils and the integrity of the endothelial cell barrier was tested in cell-based models. The in vitro IA treatment with miniaturized adsorbers resulted in reduced plasma levels of cytokines and chemokines. We also found a reduced migration of neutrophils towards patient plasma treated with the adsorbers. Furthermore, the IA-treated plasma had a positive effect on the endothelial cell barrier's integrity in the cell culture model. Our findings suggest that IA results in a reduced infiltration of cells into the central nervous system by reducing leukocyte transmigration and preventing blood-brain barrier breakdown. This novel approach of performing in vitro blood purification therapies on actual patient samples with miniaturized adsorbers and testing their effects in cell-based assays that investigate specific hypotheses of the pathophysiology provides a promising platform for elucidating the mechanisms of action of those therapies in various diseases.
Collapse
Affiliation(s)
- Andreas Körtge
- Department of Extracorporeal Therapy Systems, Fraunhofer Institute for Cell Therapy and Immunology IZI, 18057 Rostock, Germany
- Division of Nephrology, Center for Internal Medicine, Rostock University Medical Center, 18057 Rostock, Germany
| | - Anne Breitrück
- Department of Extracorporeal Therapy Systems, Fraunhofer Institute for Cell Therapy and Immunology IZI, 18057 Rostock, Germany
- Division of Nephrology, Center for Internal Medicine, Rostock University Medical Center, 18057 Rostock, Germany
| | - Sandra Doß
- Department of Extracorporeal Therapy Systems, Fraunhofer Institute for Cell Therapy and Immunology IZI, 18057 Rostock, Germany
| | - Jacqueline Hofrichter
- Department of Extracorporeal Therapy Systems, Fraunhofer Institute for Cell Therapy and Immunology IZI, 18057 Rostock, Germany
- Division of Nephrology, Center for Internal Medicine, Rostock University Medical Center, 18057 Rostock, Germany
| | - Sophie-Charlotte Nelz
- Division of Nephrology, Center for Internal Medicine, Rostock University Medical Center, 18057 Rostock, Germany
| | - Horst Krüsemann
- Department of Extracorporeal Therapy Systems, Fraunhofer Institute for Cell Therapy and Immunology IZI, 18057 Rostock, Germany
| | - Reinhold Wasserkort
- Department of Extracorporeal Therapy Systems, Fraunhofer Institute for Cell Therapy and Immunology IZI, 18057 Rostock, Germany
- Division of Nephrology, Center for Internal Medicine, Rostock University Medical Center, 18057 Rostock, Germany
| | - Brit Fitzner
- Division of Neuroimmunology, Department of Neurology, Rostock University Medical Center, 18147 Rostock, Germany
| | - Michael Hecker
- Division of Neuroimmunology, Department of Neurology, Rostock University Medical Center, 18147 Rostock, Germany
| | - Steffen Mitzner
- Department of Extracorporeal Therapy Systems, Fraunhofer Institute for Cell Therapy and Immunology IZI, 18057 Rostock, Germany
- Division of Nephrology, Center for Internal Medicine, Rostock University Medical Center, 18057 Rostock, Germany
| | - Uwe Klaus Zettl
- Division of Neuroimmunology, Department of Neurology, Rostock University Medical Center, 18147 Rostock, Germany
| |
Collapse
|
4
|
Directo D, Lee SR. Cancer Cachexia: Underlying Mechanisms and Potential Therapeutic Interventions. Metabolites 2023; 13:1024. [PMID: 37755304 PMCID: PMC10538050 DOI: 10.3390/metabo13091024] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 09/14/2023] [Accepted: 09/14/2023] [Indexed: 09/28/2023] Open
Abstract
Cancer cachexia, a multifactorial metabolic syndrome developed during malignant tumor growth, is characterized by an accelerated loss of body weight accompanied by the depletion of skeletal muscle mass. This debilitating condition is associated with muscle degradation, impaired immune function, reduced functional capacity, compromised quality of life, and diminished survival in cancer patients. Despite the lack of the known capability of fully reversing or ameliorating this condition, ongoing research is shedding light on promising preclinical approaches that target the disrupted mechanisms in the pathophysiology of cancer cachexia. This comprehensive review delves into critical aspects of cancer cachexia, including its underlying pathophysiological mechanisms, preclinical models for studying the progression of cancer cachexia, methods for clinical assessment, relevant biomarkers, and potential therapeutic strategies. These discussions collectively aim to contribute to the evolving foundation for effective, multifaceted counteractive strategies against this challenging condition.
Collapse
Affiliation(s)
| | - Sang-Rok Lee
- Department of Kinesiology, New Mexico State University, Las Cruces, NM 88003, USA;
| |
Collapse
|
5
|
Papaevangelou E, Esteves AM, Dasgupta P, Galustian C. Cyto-IL-15 synergizes with the STING agonist ADU-S100 to eliminate prostate tumors and confer durable immunity in mouse models. Front Immunol 2023; 14:1196829. [PMID: 37465665 PMCID: PMC10350564 DOI: 10.3389/fimmu.2023.1196829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/20/2023] [Indexed: 07/20/2023] Open
Abstract
Introduction Prostate cancer is one of the most commonly diagnosed malignancies in men with high mortality rates. Despite the recent therapeutic advances, such as immunotherapies, survival of patients with advance disease remains significantly low. Blockade of immune checkpoints has led to low response rates in these patients probably due to the immunosuppressive microenvironment and low mutation burden of prostate tumors. Combination of multiple immunotherapeutic regimes has also been unsatisfactory due to augmented adverse effects. To activate multiple immune-stimulatory pathways in the hostile prostate cancer microenvironment, we used a combination of cytotopically modified interleukin-15 (cyto-IL-15) with the stimulator of interferon genes (STING) agonist, ADU-S100. Methods To determine whether this combination regime could lead to both local and systemic anti-tumor effects, intratumoral administration of these agents was used in murine models of prostate cancer. Tumor growth and mouse survival were monitored, and ex vivo analyses, and RNA sequencing were performed on the tumors. Results Intratumorally injected ADU-S100 and cyto-IL-15 synergized to eliminate tumors in 58-67% of mice with unilateral tumors and promoted abscopal immunity in 50% of mice with bilateral tumors treated only at one side. Moreover, this combination regime offered immunoprotection against tumor rechallenge in 83% of cured mice. The efficacy of the combination treatment was associated with a strong innate and adaptive immune activation and induction of apoptotic and necrotic cell death. Cytokines, including type I and II interferons, and cytokine signalling pathways were activated, NK and T cell mediated cytotoxicity was increased, and B cells were activated both locally and systemically. While ADU-S100 led to an ulcerative pathology at the injection site, no other adverse effects were observed. Discussion Localised administration of a STING agonist together with cyto-IL-15 can confer significant systemic benefits and long-lasting immunity against prostate tumors while reducing immune related toxicities.
Collapse
Affiliation(s)
- Efthymia Papaevangelou
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King’s College London, Guy’s Hospital, London, United Kingdom
- Institute of Medical and Biomedical Education, St. George’s University of London, London, United Kingdom
| | - Ana M. Esteves
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Prokar Dasgupta
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King’s College London, Guy’s Hospital, London, United Kingdom
- Urology Centre, Guy’s Hospital, London, United Kingdom
| | - Christine Galustian
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King’s College London, Guy’s Hospital, London, United Kingdom
| |
Collapse
|
6
|
Di Matteo S, Munari E, Fiore PF, Santopolo S, Sampaoli C, Pelosi A, Chouaib S, Tumino N, Vacca P, Mariotti FR, Ebert S, Machwirth M, Haas D, Pezzullo M, Pietra G, Grottoli M, Buart S, Mortier E, Maggi E, Moretta L, Caruana I, Azzarone B. The roles of different forms of IL-15 in human melanoma progression. Front Immunol 2023; 14:1183668. [PMID: 37334356 PMCID: PMC10272795 DOI: 10.3389/fimmu.2023.1183668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/05/2023] [Indexed: 06/20/2023] Open
Abstract
Background Melanoma is a lethal skin cancer, and the risk of developing it is increased by exposure to ultraviolet (UV) radiation. The production of cytokines such as interleukin-15 (IL-15), induced by the exposure of skin cells to UV rays, could also promote melanoma development. The aim of this study is to investigate the possible role of Interleukin-15/Interleukin-15 Receptor α (IL-15/IL-15Rα) complexes in melanoma development. Methods The expression of IL-15/IL-15Rα complexes by melanoma cells was evaluated both ex vivo and in vitro by tissue microarray, PCR, and flow cytometry. The presence of the soluble complex (sIL-15/IL-15Rα) in the plasma of metastatic melanoma patients was detected using an ELISA assay. Subsequently, we investigated the impact of natural killer (NK) cell activation after rIL-2 starvation followed by exposure to the sIL-15/IL-15Rα complex. Finally, by analyzing public datasets, we studied the correlation between IL-15 and IL-15Rα expressions and melanoma stage, NK and T-cell markers, and overall survival (OS). Results Analysis of a melanoma tissue microarray shows a significant increase in the number of IL-15+ tumor cells from the benign nevi to metastatic melanoma stages. Metastatic melanoma cell lines express a phorbol-12-myristate-13-acetate (PMA)-cleavable membrane-bound IL-15 (mbIL-15), whereas cultures from primary melanomas express a PMA-resistant isoform. Further analysis revealed that 26% of metastatic patients present with consistently high plasmatic levels of sIL-15/IL-15Rα. When the recombinant soluble human IL-15/IL-15Rα complex is added to briefly starved rIL-2-expanded NK cells, these cells exhibit strongly reduced proliferation and levels of cytotoxic activity against K-562 and NALM-18 target cells. The analysis of public gene expression datasets revealed that high IL-15 and IL-15Rα intra-tumoral production correlates with the high levels of expression of CD5+ and NKp46+ (T and NK markers) and significantly correlates with a better OS in stages II and III, but not in stage IV. Conclusions Membrane-bound and secreted IL-15/IL-15Rα complexes are continuously present during progression in melanoma. It is notable that, although IL-15/IL-15Rα initially promoted the production of cytotoxic T and NK cells, at stage IV promotion of the development of anergic and dysfunctional cytotoxic NK cells was observed. In a subgroup of melanoma metastatic patients, the continuous secretion of high amounts of the soluble complex could represent a novel NK cell immune escape mechanism.
Collapse
Affiliation(s)
- Sabina Di Matteo
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Enrico Munari
- Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Piera Filomena Fiore
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Silvia Santopolo
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Camilla Sampaoli
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Andrea Pelosi
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Salem Chouaib
- Institut national de la santé et de la recherche médicale Unitè Mixte Rechercce (INSERM UMR) 1186, Integrative Tumor Immunology and Cancer Immunotherapy, Gustave Roussy, École Pratique des Hautes Études (EPHE), Faculty De Médecine Univ. Paris-Sud, University Paris-Saclay, Villejuif, France
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Nicola Tumino
- Immunology Research Area, Innate Lymphoid Cells Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Paola Vacca
- Immunology Research Area, Innate Lymphoid Cells Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Francesca Romana Mariotti
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Stefan Ebert
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital of Würzburg, Würzburg, Germany
| | - Markus Machwirth
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital of Würzburg, Würzburg, Germany
| | - Dorothee Haas
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital of Würzburg, Würzburg, Germany
| | - Marco Pezzullo
- Core Facility, IRCCS Bambino Gesù Children’s Hospital, Rome, Italy
| | - Gabriella Pietra
- Department of Experimental Medicine (DiMES), University of Genoa, Genoa, Italy
- Immunology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Melania Grottoli
- Immunology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Stephanie Buart
- Institut national de la santé et de la recherche médicale Unitè Mixte Rechercce (INSERM UMR) 1186, Integrative Tumor Immunology and Cancer Immunotherapy, Gustave Roussy, École Pratique des Hautes Études (EPHE), Faculty De Médecine Univ. Paris-Sud, University Paris-Saclay, Villejuif, France
| | - Erwan Mortier
- Nantes Université, Centre national de la recherche scientifique (CNRS), Inserm, CRCI2NA, Nantes, France
- LabEx IGO, Immunotherapy, Graft, Oncology, Nantes, France
| | - Enrico Maggi
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Lorenzo Moretta
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Ignazio Caruana
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital of Würzburg, Würzburg, Germany
| | - Bruno Azzarone
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| |
Collapse
|
7
|
Rodriguez-Herrera AJ, de Souza ABF, Castro TDF, Machado-Junior PA, Gomez ECM, Menezes TP, da Cruz Castro ML, Talvani A, Costa DC, Cangussú SD, Bezerra FS. Long-term e-cigarette aerosol exposure causes pulmonary emphysema in adult female and male mice. Regul Toxicol Pharmacol 2023:105412. [PMID: 37247649 DOI: 10.1016/j.yrtph.2023.105412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 05/14/2023] [Accepted: 05/21/2023] [Indexed: 05/31/2023]
Abstract
This study aimed to evaluate long-term exposure to conventional cigarette smoke (CC) and electronic cigarette (EC) aerosol in adult male and female C57BL/6 mice. Forty-eight C57BL/6 mice were used, male (n = 24) and female (n = 24), both were divided into three groups: control, CC and EC. The CC and EC groups were exposed to cigarette smoke or electronic cigarette aerosol, respectively, 3 times a day for 60 consecutive days. Afterwards, they were maintained for 60 days without exposure to cigarettes or electronic cigarette aerosol. Both cigarettes promoted an influx of inflammatory cells to the lung in males and females. All animals exposed to CC and EC showed an increase in lipid peroxidation and protein oxidation. There was an increase of IL-6 in males and females exposed to EC. The IL-13 levels were higher in the females exposed to EC and CC. Both sexes exposed to EC and CC presented tissue damage characterized by septal destruction and increased alveolar spaces compared to control. Our results demonstrated that exposure to CC and EC induced pulmonary emphysema in both sexes, and females seem to be more susceptible to EC.
Collapse
Affiliation(s)
- Andrea Jazel Rodriguez-Herrera
- Laboratory of Experimental Pathophysiology, Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto (UFOP), 35400-000, Ouro Preto, MG, Brazil
| | - Ana Beatriz Farias de Souza
- Laboratory of Experimental Pathophysiology, Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto (UFOP), 35400-000, Ouro Preto, MG, Brazil
| | - Thalles de Freitas Castro
- Laboratory of Experimental Pathophysiology, Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto (UFOP), 35400-000, Ouro Preto, MG, Brazil
| | - Pedro Alves Machado-Junior
- Laboratory of Experimental Pathophysiology, Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto (UFOP), 35400-000, Ouro Preto, MG, Brazil
| | - Elena Cecilia Marcano Gomez
- Laboratory of Experimental Pathophysiology, Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto (UFOP), 35400-000, Ouro Preto, MG, Brazil
| | - Tatiana Prata Menezes
- Laboratory of Immunobiology of Inflammation, Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto (UFOP), 35400-000, Ouro Preto, MG, Brazil
| | - Maria Laura da Cruz Castro
- Laboratory of Metabolic Biochemistry, Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto (UFOP), 35400-000, Ouro Preto, MG, Brazil
| | - André Talvani
- Laboratory of Immunobiology of Inflammation, Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto (UFOP), 35400-000, Ouro Preto, MG, Brazil
| | - Daniela Caldeira Costa
- Laboratory of Metabolic Biochemistry, Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto (UFOP), 35400-000, Ouro Preto, MG, Brazil
| | - Sílvia Dantas Cangussú
- Laboratory of Experimental Pathophysiology, Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto (UFOP), 35400-000, Ouro Preto, MG, Brazil
| | - Frank Silva Bezerra
- Laboratory of Experimental Pathophysiology, Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto (UFOP), 35400-000, Ouro Preto, MG, Brazil.
| |
Collapse
|
8
|
IL-15 Prevents the Development of T-ALL from Aberrant Thymocytes with Impaired DNA Repair Functions and Increased NOTCH1 Activation. Cancers (Basel) 2023; 15:cancers15030671. [PMID: 36765626 PMCID: PMC9913776 DOI: 10.3390/cancers15030671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 01/25/2023] Open
Abstract
We previously reported that NOD.Scid mice lacking interleukin-15 (IL-15), or IL-15 receptor alpha-chain, develop T-acute lymphoblastic leukemia (T-ALL). To understand the mechanisms by which IL-15 signaling controls T-ALL development, we studied the thymocyte developmental events in IL-15-deficient Scid mice from NOD and C57BL/6 genetic backgrounds. Both kinds of mice develop T-ALL characterized by circulating TCR-negative cells expressing CD4, CD8 or both. Analyses of thymocytes in NOD.Scid.Il15-/- mice prior to T-ALL development revealed discernible changes within the CD4-CD8- double-negative (DN) thymocyte developmental stages and increased frequencies of CD4+CD8+ double-positive cells with a high proportion of TCR-negative CD4+ and CD8+ cells. The DN cells also showed elevated expressions of CXCR4 and CD117, molecules implicated in the expansion of DN thymocytes. T-ALL cell lines and primary leukemic cells from IL-15-deficient NOD.Scid and C57BL/6.Scid mice displayed increased NOTCH1 activation that was inhibited by NOTCH1 inhibitors and blockers of the PI3K/AKT pathway. Primary leukemic cells from NOD.Scid.Il15-/- mice survived and expanded when cultured with MS5 thymic stromal cells expressing Delta-like ligand 4 and supplemented with IL-7 and FLT3 ligand. These findings suggest that IL-15 signaling in the thymus controls T-ALL development from aberrant thymocytes with an impaired DNA repair capacity and increased NOTCH1 activation.
Collapse
|
9
|
Zhou Y, Quan G, Liu Y, Shi N, Wu Y, Zhang R, Gao X, Luo L. The application of Interleukin-2 family cytokines in tumor immunotherapy research. Front Immunol 2023; 14:1090311. [PMID: 36936961 PMCID: PMC10018032 DOI: 10.3389/fimmu.2023.1090311] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
The Interleukin-2 Family contains six kinds of cytokines, namely IL-2, IL-15, IL-4, IL-7, IL-9, and IL-21, all of which share a common γ chain. Many cytokines of the IL-2 family have been reported to be a driving force in immune cells activation. Therefore, researchers have tried various methods to study the anti-tumor effect of cytokines for a long time. However, due to the short half-life, poor stability, easy to lead to inflammatory storms and narrow safety treatment window of cytokines, this field has been tepid. In recent years, with the rapid development of protein engineering technology, some engineered cytokines have a significant effect in tumor immunotherapy, showing an irresistible trend of development. In this review, we will discuss the current researches of the IL-2 family and mainly focus on the application and achievements of engineered cytokines in tumor immunotherapy.
Collapse
Affiliation(s)
- Yangyihua Zhou
- Department of Medical Laboratory, School of Medicine, Hunan Normal University, Changsha, Hunan, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Guiqi Quan
- Department of Medical Laboratory, School of Medicine, Hunan Normal University, Changsha, Hunan, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yujun Liu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Ning Shi
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
- Cancer Research Institute, Department of Neurosurgery, School of Basic Medical Science, Xiangya Hospital, Central South University, Changsha, China
| | - Yahui Wu
- Department of Medical Laboratory, School of Medicine, Hunan Normal University, Changsha, Hunan, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Ran Zhang
- Department of Medical Laboratory, School of Medicine, Hunan Normal University, Changsha, Hunan, China
- *Correspondence: Ran Zhang, ; Xiang Gao, ; Longlong Luo,
| | - Xiang Gao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
- *Correspondence: Ran Zhang, ; Xiang Gao, ; Longlong Luo,
| | - Longlong Luo
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
- *Correspondence: Ran Zhang, ; Xiang Gao, ; Longlong Luo,
| |
Collapse
|
10
|
Luo Z, He Z, Qin H, Chen Y, Qi B, Lin J, Sun Y, Sun J, Su X, Long Z, Chen S. Exercise-induced IL-15 acted as a positive prognostic implication and tumor-suppressed role in pan-cancer. Front Pharmacol 2022; 13:1053137. [PMID: 36467072 PMCID: PMC9712805 DOI: 10.3389/fphar.2022.1053137] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 11/07/2022] [Indexed: 09/12/2023] Open
Abstract
Objective: Exercise can produce a large number of cytokines that may benefit cancer patients, including Interleukin 15 (IL-15). IL-15 is a cytokine that has multiple functions in regulating the adaptive and innate immune systems and tumorigenesis of lung and breast cancers. However, the roles of IL-15 in other types of cancer remain unknown. In this article, we try to systematically analyze if IL-15 is a potential molecular biomarker for predicting patient prognosis in pan-cancer and its connection with anti-cancer effects of exercise. Methods: The expression of IL-15 was detected by The Cancer Genome Atlas (TCGA) database, Human protein Atlas (HPA), and Genotype Tissue-Expression (GTEX) database. Analysis of IL-15 genomic alterations and protein expression in human organic tissues was analyzed by the cBioPortal database and HPA. The correlations between IL-15 expression and survival outcomes, clinical features, immune-associated cell infiltration, and ferroptosis/cuproptosis were analyzed using the TCGA, ESTIMATE algorithm, and TIMER databases. Gene Set Enrichment Analysis (GSEA) was performed to evaluate the biological functions of IL-15 in pan-cancer. Results: The differential analysis suggested that the level of IL-15 mRNA expression was significantly downregulated in 12 tumor types compared with normal tissues, which is similar to the protein expression in most cancer types. The high expression of IL-15 could predict the positive survival outcome of patients with LUAD (lung adenocarcinoma), COAD (colon adenocarcinoma), COADREAD (colon and rectum adenocarcinoma), ESCA (esophageal carcinoma), SKCM (skin cutaneous melanoma), UCS (uterine carcinosarcoma), and READ (rectum adenocarcinoma). Moreover, amplification was found to be the most frequent mutation type of IL-15 genomic. Furthermore, the expression of IL-15 was correlated to the infiltration levels of various immune-associated cells in pan-cancer assessed by the ESTIMATE algorithm and TIMER database. In addition, IL-15 is positively correlated with ferroptosis/cuproptosis-related genes (ACSL4 and LIPT1) in pan-cancer. Levels of IL-15 were reported to be elevated in humans for 10-120 min following an acute exercise. Therefore, we hypothesized that the better prognosis of pan-cancer patients with regular exercise may be achieved by regulating level of IL-15. Conclusion: Our results demonstrated that IL-15 is a potential molecular biomarker for predicting patient prognosis, immunoreaction, and ferroptosis/cuproptosis in pan-cancer and partly explained the anti-cancer effects of exercise.
Collapse
Affiliation(s)
- Zhiwen Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhong He
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai, China
| | - Haocheng Qin
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai, China
| | - Yisheng Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Beijie Qi
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University, Shanghai, China
| | - Jinrong Lin
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yaying Sun
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Junming Sun
- Laboratory Animal Center, Guangxi Medical University, Nanning, China
| | - Xiaoping Su
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, College & Hospital of Stomatology, Guangxi Medical University, Nanning, China
| | - Ziwen Long
- Department of Gastric Cancer Sugery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Shiyi Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
11
|
Atallah-Yunes SA, Robertson MJ. Cytokine Based Immunotherapy for Cancer and Lymphoma: Biology, Challenges and Future Perspectives. Front Immunol 2022; 13:872010. [PMID: 35529882 PMCID: PMC9067561 DOI: 10.3389/fimmu.2022.872010] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/22/2022] [Indexed: 11/15/2022] Open
Abstract
Cytokines regulate both the innate and adaptive immune responses to cancer. Although antitumor activity has been seen for several cytokines in preclinical models, they have had limited success as single therapeutic agents in clinical trials of cancer immunotherapy. However, the possible combinations of cytokines with other immune therapeutics and the advancement in genetic engineering, synthetic biology and cellular and immune therapy has led to the revival of interest in cytokines as anticancer agents. This article will review several immunostimulatory cytokines with anticancer activity, focusing on the those that have been studied in treatment of lymphoma and highlighting recent advances of potential clinical relevance.
Collapse
Affiliation(s)
- Suheil Albert Atallah-Yunes
- Department of Hematology and Medical Oncology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Michael J Robertson
- Department of Hematology and Medical Oncology, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
12
|
Mylod E, Lysaght J, Conroy MJ. Natural killer cell therapy: A new frontier for obesity-associated cancer. Cancer Lett 2022; 535:215620. [PMID: 35283210 DOI: 10.1016/j.canlet.2022.215620] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/16/2022] [Accepted: 03/03/2022] [Indexed: 02/09/2023]
Abstract
Natural killer (NK) cell infiltration of solid tumours is associated with better outcomes, placing augmentation of NK cell abundance in tumours as an attractive immunotherapeutic approach. The unique ability of NK cells to target cancer cells without antigen specificity increases their versatility and applicability as an immunotherapeutic tool. However, successful utilisation of NK cell-based therapies in solid tumours is still at an early stage. Obesity has become a global health epidemic, and the prevalence of obesity-associated cancers has significantly increased. Obesity-associated malignancies provide a unique challenge for the successful application of cell-based immunotherapies including NK cell-based therapies because significant numbers of NK and T cells are recruited to the visceral adipose tissue at the expense of successful tumour infiltration and eradication. As such, immunotherapy efficacy has been disappointing for obesity-associated malignancies such as oesophageal and gastric adenocarcinoma. Therefore, immunotherapies for obesity-associated cancers warrant our further attention. Indeed, it is becoming ever more obvious that more innovative approaches are needed to re-invigorate anti-tumour immunity and overcome immune exclusion in such tumours. In this review, we briefly summarise the dysfunctionality of NK cells in obesity-associated cancer. We outline the NK cell-based immunotherapeutic approaches which hold promise as effective treatments in this disease space, including CAR-NK cells. Furthermore, we suggest future avenues which possess the potential to transform immunotherapy and specifically NK cell therapy efficacy for obesity-associated cancer.
Collapse
Affiliation(s)
- Eimear Mylod
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute and Trinity St. James's Cancer Institute, St. James's Hospital, Trinity College Dublin, Dublin, 8, Ireland
| | - Joanne Lysaght
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute and Trinity St. James's Cancer Institute, St. James's Hospital, Trinity College Dublin, Dublin, 8, Ireland
| | - Melissa J Conroy
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute and Trinity St. James's Cancer Institute, St. James's Hospital, Trinity College Dublin, Dublin, 8, Ireland; Cancer Immunology Research Group, Department of Physiology, School of Medicine, Trinity College Dublin, Dublin, 2, Ireland.
| |
Collapse
|
13
|
Zarrineh M, Ashrafian S, Jensen P, Nawrocki A, Ansari AM, Rezadoost H, Ghassempour A, Larsen MR. Comprehensive proteomics and sialiomics of the anti-proliferative activity of safranal on triple negative MDA-MB-231 breast cancer cell lines. J Proteomics 2022; 259:104539. [PMID: 35240313 DOI: 10.1016/j.jprot.2022.104539] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 12/27/2021] [Accepted: 02/04/2022] [Indexed: 12/17/2022]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer with no efficient treatment. Researchers have indicated the importance of quantitative approaches on proteome and different post-translation modifications studies both in diagnosis and treatment purposes. Sialic acid-containing glycopeptides (the sialiome) is one of these modifications which can be used as a tool in cancer diagnosis or therapeutic strategies since the sialylation is strongly associated with cancer migration and metastasis. Based on our study, safranal, which is a non-toxic compound in orally intakes, exhibits a significant cytotoxic effect on MDA-MB-231 in comparison to normal cells. We conducted a comprehensive proteomics and sialiomics analysis of safranal treated MDA-MB-231 cells by using a combination of TMT labeling and titanium dioxide enrichment of sialylated N-linked glycopeptides to investigate the underlying molecular mechanism behind safranal-induced apoptosis. Safranal has main effect on the inhibition of metabolism and mitochondrial dysfunction. It regulates proteins considered as activator of DNA fragmentation and apoptosis mediators. Moreover, safranal regulates sialylation of glycoproteins involving in cellular adhesion, migration and survival. It suppresses cell survival and metastasis through the alteration of the sialylation level on important signaling receptors. These results highlight the impact of safranal as a potent anticancer compound on TNBCs which also can be strongly used in daily diets. SIGNIFICANCE: In first step, we evaluated the cell viability of MDA-MB-231 cell lines against the purified saffron components (total crocin, picrocrocin, crocin I and safranal). Safranal was the only compound demonstrated the anti-proliferation effect. In order to obtain an understanding of safranal cytotoxic effect on MDA-MB-231, we designed the three set of treated cell lines in 30 min, 12 h and 24 h time-points in three replicates and a combination of TMT-based labeling quantitative proteomics and titanium dioxide (TiO2)-based enrichment of sialylated N-linked glycopeptides for sialiomics analysis as a strategy to follow the more detailed mechanisms of safranal effect. The results of bioinformatics analysis revealed the multifunction role of safranal on MDA-MB-231 cell lines. Safranal mainly dysregulates mitochondrial function, inhibits metabolism and starts initial signaling of apoptosis which lead to DNA fragmentation. Moreover, safranal caused the majority of down-regulation in sialylation profile in all time-points. Safranal also declines the cell survival, adhesion and migration by dysregulation of the sialylation level in important proteins including integrins, tumor necrosis factor receptor and cell adhesion molecules (CAMs). The results provide a set of therapeutic targets for triple negative breast cancer which can help designing of effective anticancer drugs specially in targeted therapies.
Collapse
Affiliation(s)
- Mahshid Zarrineh
- Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran 1983963113, Iran
| | - Shahrbanou Ashrafian
- Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran 1983963113, Iran
| | - Pia Jensen
- Protein research group, Department of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5230 Odense M, Denmark
| | - Arkadiusz Nawrocki
- Protein research group, Department of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5230 Odense M, Denmark
| | - Alireza Madjid Ansari
- Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Hassan Rezadoost
- Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran 1983963113, Iran
| | - Alireza Ghassempour
- Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran 1983963113, Iran.
| | - Martin R Larsen
- Protein research group, Department of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5230 Odense M, Denmark.
| |
Collapse
|
14
|
Berrien-Elliott MM, Becker-Hapak M, Cashen AF, Jacobs M, Wong P, Foster M, McClain E, Desai S, Pence P, Cooley S, Brunstein C, Gao F, Abboud CN, Uy GL, Westervelt P, Jacoby MA, Pusic I, Stockerl-Goldstein KE, Schroeder MA, DiPersio JF, Soon-Shiong P, Miller JS, Fehniger TA. Systemic IL-15 promotes allogeneic cell rejection in patients treated with natural killer cell adoptive therapy. Blood 2022; 139:1177-1183. [PMID: 34797911 PMCID: PMC9211446 DOI: 10.1182/blood.2021011532] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 11/09/2021] [Indexed: 11/20/2022] Open
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.
Collapse
Affiliation(s)
- Melissa M Berrien-Elliott
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Michelle Becker-Hapak
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Amanda F Cashen
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Miriam Jacobs
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Pamela Wong
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Mark Foster
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Ethan McClain
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Sweta Desai
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Patrick Pence
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Sarah Cooley
- Department of Medicine, University of Minnesota, Minneapolis, MN
| | | | - Feng Gao
- Department of Surgery, Washington University School of Medicine, St. Louis, MO
| | - Camille N Abboud
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Geoffrey L Uy
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Peter Westervelt
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Meagan A Jacoby
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Iskra Pusic
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | | | - Mark A Schroeder
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - John F DiPersio
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Patrick Soon-Shiong
- ImmunityBio Inc., Culver City, CA; and
- Department of Surgery, University of California, Los Angeles, CA
| | - Jeffrey S Miller
- Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Todd A Fehniger
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
15
|
Mylod E, O’Connell F, Donlon NE, Butler C, Reynolds JV, Lysaght J, Conroy MJ. The Omentum in Obesity-Associated Cancer: A Hindrance to Effective Natural Killer Cell Migration towards Tumour Which Can Be Overcome by CX3CR1 Antagonism. Cancers (Basel) 2021; 14:cancers14010064. [PMID: 35008227 PMCID: PMC8750072 DOI: 10.3390/cancers14010064] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/15/2021] [Accepted: 12/21/2021] [Indexed: 01/16/2023] Open
Abstract
Simple Summary Oesophagogastric adenocarcinomas (OAC) are cancers of the food pipe and stomach which have a strong link with obesity. Natural killer (NK) cells are assassins of the immune system and are crucial for eliminating cancer. We have shown previously that NK cells are pulled into fat in OAC patients by a signalling protein called fractalkine (CX3CL1). Once in fat, NK cells die or are profoundly altered. This diminishes their ability to kill the tumour. We report that exposure to fat can reduce movement of NK cells towards the tumour. However, if a drug called a CX3CR1 antagonist is used to antagonise the receptor for fractalkine, we can restore NK cell movement towards the tumour. When we activate NK cells with a protein called IL-15, fractalkine can reduce its effect on NK cells. This provides further evidence for using CX3CR1 antagonists to reduce NK cell migration to fat and boost NK cell movement to the tumour. Abstract Oesophagogastric adenocarcinomas (OAC) are obesity-associated malignancies, underpinned by severe immune dysregulation. We have previously shown that natural killer (NK) cells preferentially migrate to OAC omentum, where they undergo phenotypic and functional alterations and apoptosis. Furthermore, we have identified the CX3CR1:fractalkine (CX3CL1) pathway as pivotal in their recruitment to omentum. Here, we elucidate whether exposure to the soluble microenvironment of OAC omentum, and in particular fractalkine and IL-15 affects NK cell homing capacity towards oesophageal tumour. Our data uncover diminished NK cell migration towards OAC tumour tissue conditioned media (TCM) following exposure to omental adipose tissue conditioned media (ACM) and reveal that this migration can be rescued with CX3CR1 antagonist E6130. Furthermore, we show that fractalkine has opposing effects on NK cell migration towards TCM, when used alone or in combination with IL-15 and uncover its inhibitory effects on IL-15-mediated stimulation of death receptor ligand expression. Interestingly, treatment with fractalkine and/or IL-15 do not significantly affect NK cell adhesion to MAdCAM-1, despite changes they elicit to the expression of integrin α4β7. This study provides further evidence that CX3CR1 antagonism has therapeutic utility in rescuing NK cells from the deleterious effects of the omentum and fractalkine in OAC, thus limiting their dysfunction.
Collapse
Affiliation(s)
- Eimear Mylod
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James’s Hospital, Dublin 8, Ireland; (E.M.); (N.E.D.); (J.L.)
| | - Fiona O’Connell
- Department of Surgery, Trinity St. James’s Cancer Institute, Trinity College Dublin, St James’s Hospital, Dublin 8, Ireland; (F.O.); (C.B.)
| | - Noel E. Donlon
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James’s Hospital, Dublin 8, Ireland; (E.M.); (N.E.D.); (J.L.)
| | - Christine Butler
- Department of Surgery, Trinity St. James’s Cancer Institute, Trinity College Dublin, St James’s Hospital, Dublin 8, Ireland; (F.O.); (C.B.)
| | - John V. Reynolds
- Gastro-Intestinal Medicine and Surgery, St. James’s Hospital, Dublin 8, Ireland;
| | - Joanne Lysaght
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James’s Hospital, Dublin 8, Ireland; (E.M.); (N.E.D.); (J.L.)
| | - Melissa J. Conroy
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James’s Hospital, Dublin 8, Ireland; (E.M.); (N.E.D.); (J.L.)
- Cancer Immunology Research Group, Department of Physiology, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
- Correspondence:
| |
Collapse
|
16
|
Payload Delivery: Engineering Immune Cells to Disrupt the Tumour Microenvironment. Cancers (Basel) 2021; 13:cancers13236000. [PMID: 34885108 PMCID: PMC8657158 DOI: 10.3390/cancers13236000] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 02/08/2023] Open
Abstract
Although chimeric antigen receptor (CAR) T cells have shown impressive clinical success against haematological malignancies such as B cell lymphoma and acute lymphoblastic leukaemia, their efficacy against non-haematological solid malignancies has been largely disappointing. Solid tumours pose many additional challenges for CAR T cells that have severely blunted their potency, including homing to the sites of disease, survival and persistence within the adverse conditions of the tumour microenvironment, and above all, the highly immunosuppressive nature of the tumour milieu. Gene engineering approaches for generating immune cells capable of overcoming these hurdles remain an unmet therapeutic need and ongoing area of research. Recent advances have involved gene constructs for membrane-bound and/or secretable proteins that provide added effector cell function over and above the benefits of classical CAR-mediated cytotoxicity, rendering immune cells not only as direct cytotoxic effectors against tumours, but also as vessels for payload delivery capable of both modulating the tumour microenvironment and orchestrating innate and adaptive anti-tumour immunity. We discuss here the novel concept of engineered immune cells as vessels for payload delivery into the tumour microenvironment, how these cells are better adapted to overcome the challenges faced in a solid tumour, and importantly, the novel gene engineering approaches required to deliver these more complex polycistronic gene constructs.
Collapse
|
17
|
Fiore PF, Di Matteo S, Tumino N, Mariotti FR, Pietra G, Ottonello S, Negrini S, Bottazzi B, Moretta L, Mortier E, Azzarone B. Interleukin-15 and cancer: some solved and many unsolved questions. J Immunother Cancer 2021; 8:jitc-2020-001428. [PMID: 33203664 PMCID: PMC7674108 DOI: 10.1136/jitc-2020-001428] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/2020] [Indexed: 12/29/2022] Open
Abstract
Soluble interleukin (IL)-15 exists under two forms: as monomer (sIL-15) or as heterodimeric complex in association with sIL-15Rα (sIL-15/IL-15Rα). Both forms have been successfully tested in experimental tumor murine models and are currently undergoing investigation in phase I/II clinical trials. Despite more than 20 years research on IL-15, some controversial issues remain to be addressed. A first point concerns the detection of the sIL-15/IL-15Rα in plasma of healthy donors or patients with cancer and its biological significance. The second and third unsolved question regards the protumorigenic role of the IL-15/IL-15Rα complex in human cancer and the detrimental immunological consequences associated to prolonged exposure of natural killer (NK) cells to both forms of soluble IL-15, respectively. Data suggest that in vivo prolonged or repeated exposure to monomeric sIL-15 or the soluble complex may lead to NK hypo-responsiveness through the expansion of the CD8+/CD44+ T cell subset that would suppress NK cell functions. In vitro experiments indicate that soluble complex and monomeric IL-15 may cause NK hyporesponsiveness through a direct effect caused by their prolonged stimulation, suggesting that this mechanism could also be effective in vivo. Therefore, a better knowledge of IL-15 and a more appropriate use of both its soluble forms, in terms of concentrations and time of exposure, are essential in order to improve their therapeutic use. In cancer, the overproduction of sIL-15/IL-15Rα could represent a novel mechanism of immune escape. The soluble complex may act as a decoy cytokine unable to efficiently foster NK cells, or could induce NK hyporesponsiveness through an excessive and prolonged stimulation depending on the type of IL-15Rα isoforms associated. All these unsolved questions are not merely limited to the knowledge of IL-15 pathophysiology, but are crucial also for the therapeutic use of this cytokine. Therefore, in this review, we will discuss key unanswered issues on the heterogeneity and biological significance of IL-15 isoforms, analyzing both their cancer-related biological functions and their therapeutic implications.
Collapse
Affiliation(s)
| | - Sabina Di Matteo
- Immunology Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Nicola Tumino
- Immunology Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Gabriella Pietra
- Immuology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Department of Experimental Medicine (DiMES), University of Genoa, Genoa, Italy
| | - Selene Ottonello
- Department of Experimental Medicine (DiMES), University of Genoa, Genoa, Italy.,Center of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Simone Negrini
- Clinical Immunology Unit, Department of Internal Medicine, University of Genoa and Ospedale Policlinico San Martino, Genova, Italy
| | - Barbara Bottazzi
- Department of Immunology and Inflammation, Humanitas Clinical and Research Institute, Milan, Italy
| | - Lorenzo Moretta
- Immunology Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Erwan Mortier
- University of Nantes, CNRS, Inserm, CRCINA, University of Nantes, Nantes, France .,Immunotherapy, Graft, Oncology, LabEx IGO, Nantes, France
| | - Bruno Azzarone
- Immunology Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
18
|
Xu H, Shi M, Shao C, Li H, Wu J, Yu Y, Fang F, Guo Y, Xiao W. Development of IL-15/IL-15Rα sushi domain-IgG4 Fc complexes in Pichia pastoris with potent activities and prolonged half-lives. Microb Cell Fact 2021; 20:115. [PMID: 34107983 PMCID: PMC8190845 DOI: 10.1186/s12934-021-01605-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 05/31/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Interleukin-15 (IL-15) is a critical cytokine for the development, proliferation, and function of natural killer (NK) cells, NKT cells, and CD8+ memory T cells and has become one of the most promising protein molecules for the treatment of cancer and viral diseases. However, there are several limitations in applying IL-15 in therapy, such as its low yield in vitro, limited potency, and short half-life in vivo. To date, there are several recombinant IL-15 agonists based on configurational modifications that are being pursued in the treatment of cancer, such as ALT-803, which are mainly produced from mammalian cells. RESULTS In this study, we designed two different forms of the IL-15 complex, which were formed by the noncovalent assembly of IL-15 with dimeric or monomeric sushi domain of IL-15 receptor α (SuIL-15Rα)-IgG4 Fc fusion protein and designated IL-15/SuIL-15Rα-dFc and IL-15/SuIL-15Rα-mFc, respectively. The two IL-15 complexes were expressed in Pichia pastoris (P. pastoris), and their activities and half-lives were evaluated and compared. Pharmacokinetic analysis showed that IL-15/SuIL-15Rα-dFc had a half-life of 14.26 h while IL-15/SuIL-15Rα-mFc had a half-life of 9.16 h in mice, which were much longer than the 0.7-h half-life of commercial recombinant human IL-15 (rhIL-15). Treatment of mice with intravenous injection of the two IL-15 complexes resulted in significant increases in NK cells, NKT cells, and memory CD8+ T cells, which were not observed after rhIL-15 treatment. Treatment of human peripheral blood mononuclear cells (PBMCs) from healthy donors with the two IL-15 complexes yielded enhanced NK and CD8+ T cell activation and proliferation, which was comparable to the effect of rhIL-15. CONCLUSIONS These findings indicate that the IL-15/SuIL-15Rα-dFc and IL-15/SuIL-15Rα-mFc produced in P. pastoris exhibit potent activities and prolonged half-lives and may serve as superagonists for immunotherapy in further research and applications.
Collapse
Affiliation(s)
- Huan Xu
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China.,Institute of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China.,Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Mingyang Shi
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China.,Institute of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China.,Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Changsheng Shao
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China.,Institute of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China.,Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Hao Li
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China.,Institute of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China.,Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Jing Wu
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China.,Institute of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China.,Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Yin Yu
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China.,Institute of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China.,Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Fang Fang
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China.,Institute of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China.,Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Yugang Guo
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China. .,Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China. .,Institute of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China. .,Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei, 230027, Anhui, China.
| | - Weihua Xiao
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China. .,Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China. .,Institute of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China. .,Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei, 230027, Anhui, China.
| |
Collapse
|
19
|
Isvoranu G, Surcel M, Munteanu AN, Bratu OG, Ionita-Radu F, Neagu MT, Chiritoiu-Butnaru M. Therapeutic potential of interleukin-15 in cancer (Review). Exp Ther Med 2021; 22:675. [PMID: 33986840 PMCID: PMC8112152 DOI: 10.3892/etm.2021.10107] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 02/24/2021] [Indexed: 12/21/2022] Open
Abstract
The immune system is dysfunctional in cancer, and therapeutic approaches designated to restore immunity and increase long-term overall survival are desirable. The role of immunotherapy is to trigger the immune system to recognize and destroy tumor cells. Interleukin-15 (IL-15) is a member of the common gamma-chain (γc) cytokines that promote the differentiation and expansion of T cells, B cells and natural killer (NK) cells, leading to enhanced antitumor responses. This suggests that IL-15 is a promising candidate for anticancer therapy. Renewed interest in cancer immunotherapy has led to an increased number of preclinical studies and clinical trials that have investigated the reliability and potency of IL-15-based agents, not only as single therapy, but also in combination with others. This review provides a description of these studies which show the advantages and disadvantages of IL-15 as an immunotherapeutic agent. We present here the role of IL-15 and pharmacologically improved IL-15 superagonists as a single treatment or in combination with other therapeutic agents.
Collapse
Affiliation(s)
- Gheorghita Isvoranu
- Department of Animal Husbandry, 'Victor Babes' National Institute of Pathology, 050096 Bucharest, Romania
| | - Mihaela Surcel
- Department of Immunology, 'Victor Babes' National Institute of Pathology, 050096 Bucharest, Romania
| | - Adriana Narcisa Munteanu
- Department of Immunology, 'Victor Babes' National Institute of Pathology, 050096 Bucharest, Romania.,Department of Doctoral School of Biology, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania
| | - Ovidiu Gabriel Bratu
- Department of Clinical Department III, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania.,Department of Clinic of Urology, 'Dr. Carol Davila' University Emergency Central Military Hospital, 010825 Bucharest, Romania.,Academy of Romanian Scientists, 050094 Bucharest, Romania
| | - Florentina Ionita-Radu
- Department of Gastroenterology, 'Dr. Carol Davila' University Emergency Central Military Hospital, 010825 Bucharest, Romania
| | - Monica Teodora Neagu
- Department of Immunology, 'Victor Babes' National Institute of Pathology, 050096 Bucharest, Romania.,Department of Doctoral School of Biology, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania
| | - Marioara Chiritoiu-Butnaru
- Department of Molecular Cell Biology, Institute of Biochemistry of the Romanian Academy (IBAR), 060031 Bucharest, Romania
| |
Collapse
|
20
|
D'mello KP, Zhao L, Kaser EC, Zhu Z, Xiao H, Wakefield MR, Bai Q, Fang Y. The role of interleukins and the widely studied TNF-α in non-Hodgkin's lymphoma. Med Oncol 2021; 38:56. [PMID: 33835307 DOI: 10.1007/s12032-021-01504-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 03/27/2021] [Indexed: 11/25/2022]
Abstract
Non-Hodgkin's lymphoma is a relatively common cancer with malignant tendencies. Although there is no current cure for the disease, research has been successful in figuring out the mechanisms of how the disease progresses, however, there is still a lot of unknowns, and more research must be done to find the ultimate cause of the disease. Interleukins and various cytokines play unique roles in the development of cancer. This review article summarizes the pathophysiology between certain interleukins and non-Hodgkin's lymphoma. Although TNF-α is not an interleukin, the article examines TNF-α due to its high correspondence with cancer. The article also describes a promising immunotherapy for the disease, while looking at immunotherapies that have been successful in other types of cancer and disease.
Collapse
Affiliation(s)
- Kyle P D'mello
- Department of Microbiology, Immunology and Pathology, Des Moines University College of Osteopathic Medicine, Des Moines, IA, 50312, USA
| | - Lei Zhao
- The Department of Respiratory Medicine, The 2nd People's Hospital of Hefei and Hefei Hospital Affiliated to Anhui Medical University, Hefei, China
| | - Erin C Kaser
- Department of Microbiology, Immunology and Pathology, Des Moines University College of Osteopathic Medicine, Des Moines, IA, 50312, USA
| | - Ziwen Zhu
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Huaping Xiao
- Department of Microbiology, Immunology and Pathology, Des Moines University College of Osteopathic Medicine, Des Moines, IA, 50312, USA.,The Affiliated Hospital of Xiangnan University, Chenzhou, Hunan, China
| | - Mark R Wakefield
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Qian Bai
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Yujiang Fang
- Department of Microbiology, Immunology and Pathology, Des Moines University College of Osteopathic Medicine, Des Moines, IA, 50312, USA. .,Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA.
| |
Collapse
|
21
|
Wang X, Zhao XY. Transcription Factors Associated With IL-15 Cytokine Signaling During NK Cell Development. Front Immunol 2021; 12:610789. [PMID: 33815365 PMCID: PMC8013977 DOI: 10.3389/fimmu.2021.610789] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 03/01/2021] [Indexed: 12/16/2022] Open
Abstract
Natural killer (NK) cells are lymphocytes primarily involved in innate immunity and possess important functional properties in anti-viral and anti-tumor responses; thus, these cells have broad potential for clinical utilization. NK cells originate from hematopoietic stem cells (HSCs) through the following two independent and continuous processes: early commitment from HSCs to IL-15-responsive NK cell progenitors (NKPs) and subsequent differentiation into mature NK cells in response to IL-15. IL-15 is the most important cytokine for NK cell development, is produced by both hematopoietic and nonhematopoietic cells, and functions through a distinct delivery process termed transpresentation. Upon being transpresented to NK cells, IL-15 contributes to NK cell development via the activation of several downstream signaling pathways, including the Ras-MEK-MAPK, JAK-STAT5, and PI3K-ATK-mTOR pathways. Nonetheless, the exact role of IL-15 in NK cell development has not been discussed in a consecutive and comprehensive manner. Here, we review current knowledge about the indispensable role of IL-15 in NK cell development and address which cells produce IL-15 to support NK cell development and when IL-15 exerts its function during multiple developmental stages. Specifically, we highlight how IL-15 supports NK cell development by elucidating the distinct transpresentation of IL-15 to NK cells and revealing the downstream target of IL-15 signaling during NK cell development.
Collapse
Affiliation(s)
- Xiang Wang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Xiang-Yu Zhao
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Beijing, China.,Beijing Engineering Laboratory for Cellular Therapy, Beijing, China
| |
Collapse
|
22
|
Corbellari R, Stringhini M, Mock J, Ongaro T, Villa A, Neri D, De Luca R. A Novel Antibody-IL15 Fusion Protein Selectively Localizes to Tumors, Synergizes with TNF-based Immunocytokine, and Inhibits Metastasis. Mol Cancer Ther 2021; 20:859-871. [PMID: 33632875 DOI: 10.1158/1535-7163.mct-20-0853] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 01/14/2021] [Accepted: 02/09/2021] [Indexed: 12/23/2022]
Abstract
IL15 is an immunostimulatory cytokine that holds promises for cancer therapy, but its performance (alone or as partner for fusion proteins) has often been limited by suboptimal accumulation in the tumor and very rapid clearance from circulation. Most recently, the Sushi Domain (SD, the shortest region of IL15 receptor α, capable of binding to IL15) has been fused to IL15-based anticancer products to increase its biological activity. Here, we describe two novel antibody fusion proteins (termed F8-F8-IL15 and F8-F8-SD-IL15), specific to the alternatively spliced EDA domain of fibronectin (a marker of tumor neoangiogenisis, expressed in the majority of solid and hematologic tumors, but absent in normal healthy tissues) and featuring the F8 antibody in single-chain diabody format (with a short linker between VH and VL, thus allowing the domains to pair with the complementary ones of another chain). Unlike previously described fusions of the F8 antibody with human IL15, F8-F8-IL15 and F8-F8-SD-IL15 exhibited a preferential uptake in solid tumors, as evidenced by quantitative biodistribution analysis with radioiodinated protein preparations. Both products were potently active in vivo against mouse metastatic colon carcinomas and in sarcoma lesion in combination with targeted TNF. The results may be of clinical significance, as F8-F8-IL15 and F8-F8-SD-IL15 are fully human proteins, which recognize the cognate tumor-associated antigen with identical affinity in mouse and man.
Collapse
Affiliation(s)
- Riccardo Corbellari
- CiBIO (Department of Cellular, Computational and Integrative Biology), University of Trento, Povo, Trento, Italy.,Philochem AG, Otelfingen, Switzerland
| | - Marco Stringhini
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | - Jaqueline Mock
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | | | | | - Dario Neri
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | | |
Collapse
|
23
|
Liao YM, Hung TH, Tung JK, Yu J, Hsu YL, Hung JT, Yu AL. Low Expression of IL-15 and NKT in Tumor Microenvironment Predicts Poor Outcome of MYCN-Non-Amplified Neuroblastoma. J Pers Med 2021; 11:jpm11020122. [PMID: 33668573 PMCID: PMC7918138 DOI: 10.3390/jpm11020122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 12/11/2022] Open
Abstract
Immune tumor microenvironment (TME) in neuroblastoma (NBL) contributes to tumor behavior and treatment response. T cells and natural killer (NK) cells have been shown to play important roles in the neuroblastoma TME. However, few reports address the clinical relevance of natural killer T cells (NKTs) and interleukin-15 (IL-15), one of the crucial cytokines controlling the activation and expansion of NK/NKT cells, in NBL. In this study, we examined NKT immunoscores and IL-15 expression in both MYCN-amplified and MYCN-non-amplified NBL to correlate with clinical outcomes such as event-free survival (EFS) and overall survival (OS). From Gene Expression Omnibus (GEO) datasets GSE45480 (n = 643) and GSE49711 (n = 493), we found that NKT immunoscore and IL-15 expression were both significantly lower in MYCN-amplified NBL, and similar results were observed using our clinical NBL samples (n = 53). Moreover, NBL patients (GEO dataset GSE49711 and our clinical samples) with both lower NKT immunoscore and IL-15 expression exhibited decreased EFS and OS regardless of MYCN gene amplification status. Multivariate analysis further showed that the combination of low NKT immunoscore and low IL-15 expression level was an independent prognostic factor for poor EFS and OS in our NBL patients. These findings provide the rationale for the development of strategy to incorporate IL-15 and NKT cell therapy into the treatment regimen for neuroblastoma.
Collapse
Affiliation(s)
- Yu-Mei Liao
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan 333, Taiwan; (Y.-M.L.); (T.-H.H.); (J.K.T.); (J.Y.)
- Program in Translational Medicine, Kaohsiung Medical University, Kaohsiung, and Academia Sinica, Taipei 115, Taiwan
- Division of Hematology and Oncology, Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Tsai-Hsien Hung
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan 333, Taiwan; (Y.-M.L.); (T.-H.H.); (J.K.T.); (J.Y.)
| | - John K. Tung
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan 333, Taiwan; (Y.-M.L.); (T.-H.H.); (J.K.T.); (J.Y.)
| | - John Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan 333, Taiwan; (Y.-M.L.); (T.-H.H.); (J.K.T.); (J.Y.)
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 115, Taiwan
| | - Ya-Ling Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Jung-Tung Hung
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan 333, Taiwan; (Y.-M.L.); (T.-H.H.); (J.K.T.); (J.Y.)
- Correspondence: (J.-T.H.); (A.L.Y.); Tel.: +886-3328-1200 (ext. 7813) (J.-T.H.); +886-3328-1200 (ext. 7805) (A.L.Y.); Fax: +886-3328-1200 (A.L.Y. & J.-T.H.)
| | - Alice L. Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan 333, Taiwan; (Y.-M.L.); (T.-H.H.); (J.K.T.); (J.Y.)
- Department of Pediatrics, University of California in San Diego, San Diego, CA 92103, USA
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
- Correspondence: (J.-T.H.); (A.L.Y.); Tel.: +886-3328-1200 (ext. 7813) (J.-T.H.); +886-3328-1200 (ext. 7805) (A.L.Y.); Fax: +886-3328-1200 (A.L.Y. & J.-T.H.)
| |
Collapse
|
24
|
Van Audenaerde JR, Marcq E, von Scheidt B, Davey AS, Oliver AJ, De Waele J, Quatannens D, Van Loenhout J, Pauwels P, Roeyen G, Lardon F, Slaney CY, Peeters M, Kershaw MH, Darcy PK, Smits EL. Novel combination immunotherapy for pancreatic cancer: potent anti-tumor effects with CD40 agonist and interleukin-15 treatment. Clin Transl Immunology 2020; 9:e1165. [PMID: 32821382 PMCID: PMC7428816 DOI: 10.1002/cti2.1165] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 07/08/2020] [Accepted: 07/20/2020] [Indexed: 12/16/2022] Open
Abstract
Objectives With the poorest 5‐year survival of all cancers, improving treatment for pancreatic cancer is one of the biggest challenges in cancer research. We sought to explore the potential of combining both priming and activation of the immune system. To achieve this, we combined a CD40 agonist with interleukin‐15 and tested its potential in pancreatic cancer. Methods Response to this combination regimen was assessed in pancreatic ductal adenocarcinoma mouse models, and a thorough analysis of the tumor microenvironment was performed. Results We demonstrated profound reduction in tumor growth and increased survival of mice with the majority of mice being cured when both agents were combined, including an unprecedented 8‐fold dose reduction of CD40 agonist without losing any efficacy. RNAseq analysis showed involvement of natural killer (NK) cell‐ and T‐cell‐mediated anti‐tumor responses and the importance of antigen‐presenting cell pathways. This combination resulted in enhanced infiltration of tumors by both T cells and NK cells, as well as a striking increase in the ratio of CD8+ T cells over Tregs. We also observed a significant increase in numbers of dendritic cells (DCs) in tumor‐draining lymph nodes, particularly CD103+ DCs with cross‐presentation potential. A critical role for CD8+ T cells and involvement of NK cells in the anti‐tumor effect was highlighted. Importantly, strong immune memory was established, with an increase in memory CD8+ T cells only when both interleukin‐15 and the CD40 agonist were combined. Conclusion These novel preclinical data support initiation of a first‐in‐human clinical trial with this combination immunotherapy strategy in pancreatic cancer.
Collapse
Affiliation(s)
- Jonas Rm Van Audenaerde
- Center for Oncological Research (CORE) Integrated Personalized & Precision Oncology Network (IPPON) University of Antwerp Wilrijk Belgium.,Cancer Immunotherapy and Immune Innovation Laboratory Peter MacCallum Cancer Centre Melbourne VIC Australia
| | - Elly Marcq
- Center for Oncological Research (CORE) Integrated Personalized & Precision Oncology Network (IPPON) University of Antwerp Wilrijk Belgium
| | - Bianca von Scheidt
- Cancer Immunotherapy and Immune Innovation Laboratory Peter MacCallum Cancer Centre Melbourne VIC Australia
| | - Ashleigh S Davey
- Cancer Immunotherapy and Immune Innovation Laboratory Peter MacCallum Cancer Centre Melbourne VIC Australia
| | - Amanda J Oliver
- Cancer Immunotherapy and Immune Innovation Laboratory Peter MacCallum Cancer Centre Melbourne VIC Australia
| | - Jorrit De Waele
- Center for Oncological Research (CORE) Integrated Personalized & Precision Oncology Network (IPPON) University of Antwerp Wilrijk Belgium
| | - Delphine Quatannens
- Center for Oncological Research (CORE) Integrated Personalized & Precision Oncology Network (IPPON) University of Antwerp Wilrijk Belgium
| | - Jinthe Van Loenhout
- Center for Oncological Research (CORE) Integrated Personalized & Precision Oncology Network (IPPON) University of Antwerp Wilrijk Belgium
| | - Patrick Pauwels
- Center for Oncological Research (CORE) Integrated Personalized & Precision Oncology Network (IPPON) University of Antwerp Wilrijk Belgium.,Department of Pathology Antwerp University Hospital Edegem Belgium
| | - Geert Roeyen
- Department of Hepatobiliary, Endocrine and Transplantation Surgery Antwerp University Hospital Edegem Belgium
| | - Filip Lardon
- Center for Oncological Research (CORE) Integrated Personalized & Precision Oncology Network (IPPON) University of Antwerp Wilrijk Belgium
| | - Clare Y Slaney
- Cancer Immunotherapy and Immune Innovation Laboratory Peter MacCallum Cancer Centre Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology The University of Melbourne Parkville VIC Australia
| | - Marc Peeters
- Center for Oncological Research (CORE) Integrated Personalized & Precision Oncology Network (IPPON) University of Antwerp Wilrijk Belgium.,Department of Oncology and Multidisciplinary Oncological Centre Antwerp Antwerp University Hospital Edegem Belgium
| | - Michael H Kershaw
- Cancer Immunotherapy and Immune Innovation Laboratory Peter MacCallum Cancer Centre Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology The University of Melbourne Parkville VIC Australia
| | - Phillip K Darcy
- Cancer Immunotherapy and Immune Innovation Laboratory Peter MacCallum Cancer Centre Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology The University of Melbourne Parkville VIC Australia
| | - Evelien Ljm Smits
- Center for Oncological Research (CORE) Integrated Personalized & Precision Oncology Network (IPPON) University of Antwerp Wilrijk Belgium.,Center for Cell Therapy and Regenerative Medicine Antwerp University Hospital Edegem Belgium
| |
Collapse
|
25
|
Waldmann TA, Miljkovic MD, Conlon KC. Interleukin-15 (dys)regulation of lymphoid homeostasis: Implications for therapy of autoimmunity and cancer. J Exp Med 2020; 217:132622. [PMID: 31821442 PMCID: PMC7037239 DOI: 10.1084/jem.20191062] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 08/30/2019] [Accepted: 10/30/2019] [Indexed: 12/15/2022] Open
Abstract
IL-15 supports NK, NK-T, γδ, ILC1, and memory CD8 T cell function, and dysregulated IL-15 is associated with many autoimmune diseases. Striking IL-15–driven increases in NK and CD8 T cells in patients highlight the potential for combination therapy of cancers. IL-15, a pleiotropic cytokine, stimulates generation of NK, NK-T, γδ, ILC1, and memory CD8 T cells. IL-15 disorders play pathogenetic roles in organ-specific autoimmune diseases including celiac disease. Diverse approaches are developed to block IL-15 action. IL-15 administered to patients with malignancy yielded dramatic increases in NK numbers and modest increases in CD8 T cells. Due to immunological checkpoints, to achieve major cancer therapeutic efficacy, IL-15 will be used in combination therapy, and combination trials with checkpoint inhibitors, with anti-CD40 to yield tumor-specific CD8 T cells, and with anticancer monoclonal antibodies to increase ADCC and antitumor efficacy, have been initiated.
Collapse
Affiliation(s)
- Thomas A Waldmann
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Milos D Miljkovic
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Kevin C Conlon
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
26
|
IL-15 negatively regulates curdlan-induced IL-23 production by human monocyte-derived dendritic cells and subsequent Th17 response. North Clin Istanb 2020; 6:379-387. [PMID: 31909384 PMCID: PMC6936942 DOI: 10.14744/nci.2019.38802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 08/19/2019] [Indexed: 11/20/2022] Open
Abstract
OBJECTIVE: In this study, we aimed to assess the effects of long- and short-term IL-15 cytokine exposure of human monocyte-derived curdlan-matured dendritic cells (DCs) on the production of Th17 cell-polarizing cytokine IL-23 and subsequent Th17 cell activation. METHODS: Peripheral blood mononuclear cells (PBMCs) were purified using Ficoll-Paque from healthy donors. Monocytes were magnetically selected using CD14 Miltenyi beads and differentiated into DCs with granulocyte-macrophage colony-stimulating factor (GM-CSF) and IL-4 for five days in the presence or absence of IL-15 (100ng/ml) for long-term exposure experiments. Then, DCs were matured with peptidoglycan (PGN), or curdlan for 24 hours. For short-term exposure experiments, IL-15 was added only during maturation of DCs. Then, DCs were characterized concerning the expression of MHC II and costimulatory molecules, production of cytokine subunits IL-23p19, IL-12p40, IL-12p35 and cytokine IL-23 via flow cytometry or real-time qPCR or ELISA. Finally, the phosphorylation of signaling molecules after curdlan stimulation was assessed using phospho-flow assays. RESULTS: IL-15 exposure suppressed IL-23 production by DCs. As a result, IL-15-exposed DCs suppressed IL-17 production by allogeneic T cells. Importantly, we observed a reduction in the surface Dectin-1 receptor levels by IL-15-exposed DCs. In line with these observations, curdlan stimulation resulted in reduced phosphorylation of ERK1/2, NF-kB p65 and AKT by human DCs exposed to IL-15 compared with controls. These results may explain why IL-15-exposed DCs produce less IL-23 after maturation with curdlan, which is a ligand of Dectin-1. CONCLUSION: Short- or long-term exposure to IL-15 of human DCs during their differentiation or maturation programs DCs against Th17 cell polarization, which suggests that IL-15 availability may affect CD4+ T cell-mediated protective immunity to fungal infections.
Collapse
|
27
|
Sakellariou C, Elhage O, Papaevangelou E, Giustarini G, Esteves AM, Smolarek D, Smith RA, Dasgupta P, Galustian C. Prostate cancer cells enhance interleukin-15-mediated expansion of NK cells. BJU Int 2019; 125:89-102. [PMID: 31392791 DOI: 10.1111/bju.14893] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVES To identify cytokines that can activate and expand NK cells in the presence of prostate cancer cells in order to determine whether these agents may be useful in future intra-tumoural administration in pre-clinical and clinical prostate cancer trials. MATERIALS AND METHODS Lymphocytes isolated from normal donor blood were set up in co-cultures with either cancer or non-cancerous prostate cell lines, together with each of the cytokines interleukin (IL)-2, IL-12, IL-15, interferon (IFN)-γ or IL-21 for a period of 7 days. Then, expansion of NK cells, NKT cells and CD8 T cells was measured by flow cytometry and compared with the expansion of the same cells in the absence of prostate cells. The cytotoxic activity of NK cells, as measured by perforin and tumour cell killing, was also assessed. NK cell receptors and their corresponding ligands on prostate tumour cells were analysed to determine whether any of these were modulated by co-culture. The role of the tumour-secreted heat shock proteins HSP90 and HSP70 in the expansion of NK cells in the co-cultures was also investigated because of their effects on NK and CD8 T-cell activation. RESULTS We showed that, among a panel of cytokines known to cause NK cell activation and expansion, only IL-15 could actively induce expansion of NK, NKT and CD8 T cells in the presence of prostate cancer cell lines. Furthermore, the expansion of NK cells was far greater (up to 50% greater) in the presence of the cancer cells (LNCaP, PC3) than when lymphocytes were incubated alone. In contrast, non-cancerous cell lines (PNT2 and WPMY-1) did not exert any expansion of NK cells. The cytolytic activity of the NK cells, as measured by perforin, CD107a and killing of tumour cells, was also greatest in co-cultures with IL-15. Examination of NK cell receptors shows that NKG2D is upregulated to a greater degree in the presence of prostate cancer cells, compared with the upregulation with IL-15 in lymphocytes alone. However, blocking of NKG2D does not inhibit the enhanced expansion of NK cells in the presence of tumour cells. CONCLUSIONS Among a panel of NK cell-activating cytokines, IL-15 was the only cytokine that could stimulate expansion of NK cells in the presence of prostate cancer cells; therefore IL-15 may be a good candidate for novel future intra-tumoural therapy of the disease.
Collapse
Affiliation(s)
- Christina Sakellariou
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Kings College London, Guys Hospital, London, UK
| | | | - Efthymia Papaevangelou
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Kings College London, Guys Hospital, London, UK
| | - Giulio Giustarini
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Kings College London, Guys Hospital, London, UK
| | - Ana M Esteves
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Kings College London, Guys Hospital, London, UK
| | - Dorota Smolarek
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Kings College London, Guys Hospital, London, UK
| | - Richard A Smith
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Kings College London, Guys Hospital, London, UK
| | - Prokar Dasgupta
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Kings College London, Guys Hospital, London, UK.,Urology Centre, Guys Hospital, London, UK
| | - Christine Galustian
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Kings College London, Guys Hospital, London, UK
| |
Collapse
|
28
|
Hennessy RJ, Pham K, Delconte R, Rautela J, Hodgkin PD, Huntington ND. Quantifying NK cell growth and survival changes in response to cytokines and regulatory checkpoint blockade helps identify optimal culture and expansion conditions. J Leukoc Biol 2019; 105:1341-1354. [PMID: 31079418 DOI: 10.1002/jlb.ma0718-296r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 04/01/2019] [Accepted: 04/28/2019] [Indexed: 01/02/2023] Open
Abstract
NK cells are innate lymphocytes critical for immune surveillance, particularly in eradication of metastatic cancer cells and acute antiviral responses. In contrast to T cells, NK cell-mediated immunity is rapid, with spontaneous cytotoxicity and cytokine/chemokine production upon pathogen detection. The renaissance in cancer immunology has cast NK cell biology back into the spotlight with an urgent need for deeper understanding of the regulatory networks that govern NK cell antitumor activity. To this end, we have adapted and refined a series of quantitative cellular calculus methods, previously applied to T and B lymphocytes, to dissect the biologic outcomes of NK cells following stimulation with cytokines (IL-15, IL-12, IL-18) or deletion of genes that regulate NK cell proliferation (Cish), survival (Bcl2l11), and activation-induced-cell-death (AICD; Fas). Our methodology is well suited to delineate effects on division rate, intrinsic apoptosis, and AICD, permitting variables such as population half-life, rate of cell division, and their combined influence on population numbers in response to stimuli to be accurately measured and modelled. Changes in these variables that result from gene deletion, concentration of stimuli, time, and cell density give insight into the dynamics of NK cell responses and serve as a platform to dissect the mechanism of action of putative checkpoints in NK cell activation and novel NK cell immunotherapy agents.
Collapse
Affiliation(s)
- Robert J Hennessy
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Kim Pham
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Rebecca Delconte
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Jai Rautela
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
- Biomedicine Discovery Institute and the Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Philip D Hodgkin
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Nicholas D Huntington
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| |
Collapse
|
29
|
Lérias JR, Paraschoudi G, Silva I, Martins J, de Sousa E, Condeço C, Figueiredo N, Carvalho C, Dodoo E, Jäger E, Rao M, Maeurer M. Clinically Relevant Immune Responses against Cytomegalovirus: Implications for Precision Medicine. Int J Mol Sci 2019; 20:ijms20081986. [PMID: 31018546 PMCID: PMC6514820 DOI: 10.3390/ijms20081986] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 04/16/2019] [Accepted: 04/16/2019] [Indexed: 12/12/2022] Open
Abstract
Immune responses to human cytomegalovirus (CMV) can be used to assess immune fitness in an individual. Further to its clinical significance in posttransplantation settings, emerging clinical and translational studies provide examples of immune correlates of protection pertaining to anti-CMV immune responses in the context of cancer or infectious diseases, e.g., tuberculosis. In this viewpoint, we provide a brief overview about CMV-directed immune reactivity and immune fitness in a clinical context and incorporate some of our own findings obtained from peripheral blood or tumour-infiltrating lymphocytes (TIL) from patients with advanced cancer. Observations in patients with solid cancers whose lesions contain both CMV and tumour antigen-specific T-cell subsets are highlighted, due to a possible CMV-associated “bystander” effect in amplifying local inflammation and subsequent tumour rejection. The role of tumour-associated antibodies recognising diverse CMV-derived epitopes is also discussed in light of anti-cancer immune responses. We discuss here the use of anti-CMV immune responses as a theranostic tool—combining immunodiagnostics with a personalised therapeutic potential—to improve treatment outcomes in oncological indications.
Collapse
Affiliation(s)
- Joana R Lérias
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Av. Brasília, 1400-038 Lisbon, Portugal.
| | - Georgia Paraschoudi
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Av. Brasília, 1400-038 Lisbon, Portugal.
| | - Inês Silva
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Av. Brasília, 1400-038 Lisbon, Portugal.
| | - João Martins
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Av. Brasília, 1400-038 Lisbon, Portugal.
| | - Eric de Sousa
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Av. Brasília, 1400-038 Lisbon, Portugal.
| | - Carolina Condeço
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Av. Brasília, 1400-038 Lisbon, Portugal.
| | - Nuno Figueiredo
- Digestive Unit, Champalimaud Centre for the Unknown, Av. Brasília, 1400-038 Lisbon, Portugal.
| | - Carlos Carvalho
- Digestive Unit, Champalimaud Centre for the Unknown, Av. Brasília, 1400-038 Lisbon, Portugal.
| | - Ernest Dodoo
- Department of Oncology and Haematology, Krankenhaus Nordwest, Steinbacher Hohl 2-26, 60488 Frankfurt am Main, Germany.
| | - Elke Jäger
- Department of Oncology and Haematology, Krankenhaus Nordwest, Steinbacher Hohl 2-26, 60488 Frankfurt am Main, Germany.
| | - Martin Rao
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Av. Brasília, 1400-038 Lisbon, Portugal.
| | - Markus Maeurer
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Av. Brasília, 1400-038 Lisbon, Portugal.
- Department of Oncology and Haematology, Krankenhaus Nordwest, Steinbacher Hohl 2-26, 60488 Frankfurt am Main, Germany.
| |
Collapse
|
30
|
The characteristics of vessel lining cells in normal spleens and their role in the pathobiology of myelofibrosis. Blood Adv 2019; 2:1130-1145. [PMID: 29776986 DOI: 10.1182/bloodadvances.2017015073] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 03/13/2018] [Indexed: 12/31/2022] Open
Abstract
The CD34-CD8α+, sinusoid lining, littoral cells (LCs), and CD34+CD8α-, splenic vascular endothelial cells (SVECs) represent 2 distinct cellular types that line the vessels within normal spleens and those of patients with myelofibrosis (MF). To further understand the respective roles of LCs and SVECs, each was purified from normal and MF spleens, cultured, and characterized. Gene expression profiling indicated that LCs were a specialized type of SVEC. LCs possessed a distinct gene expression profile associated with cytoskeleton regulation, cellular interactions, endocytosis, and iron transport. LCs also were characterized by strong phagocytic activity, less robust tube-forming capacity and a limited proliferative potential. These characteristics underlie the role of LCs as cellular filters and scavengers. Although normal LCs and SVECs produced overlapping as well as distinct hematopoietic factors and adhesion molecules, the gene expression profile of MF LCs and SVECs distinguished them from their normal counterparts. MF SVECs were characterized by activated interferon signaling and cell cycle progression pathways and increased vascular endothelial growth factor receptor, angiopoietin-2, stem cell factor, interleukin (IL)-33, Notch ligands, and IL-15 transcripts. In contrast, the transcription profile of MF LCs was associated with mitochondrial dysfunction, reduced energy production, protein biosynthesis, and catabolism. Normal SVECs formed in vitro confluent cell layers that supported MF hematopoietic colony formation to a greater extent than normal colony formation. These data provide an explanation for the reduced density of LCs observed within MF spleens and indicate the role of SVECs in the development of extramedullary hematopoiesis in MF.
Collapse
|
31
|
Zhu L, Kalimuthu S, Oh JM, Gangadaran P, Baek SH, Jeong SY, Lee SW, Lee J, Ahn BC. Enhancement of antitumor potency of extracellular vesicles derived from natural killer cells by IL-15 priming. Biomaterials 2018; 190-191:38-50. [PMID: 30391801 DOI: 10.1016/j.biomaterials.2018.10.034] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/26/2018] [Accepted: 10/26/2018] [Indexed: 12/12/2022]
Abstract
PURPOSE Natural killer (NK) cells are the key subset of innate-immunity lymphocytes; they possess antitumor activities and are used for cancer immunotherapy. In a previous study, extracellular vehicles (EVs) from NK-92MI cells were isolated and exploited for their ability to kill human cancer cells in vitro and in vivo (multiple injection methods). Here, the potential of NK-cell-derived EVs (NK-EVs) for immunotherapy was improved by priming with interleukin (IL)-15. METHODS NK-EVs were isolated from the culture medium without or with IL-15 (NK-EVsIL-15) by ultracentrifugation and were purified via density gradient ultracentrifugation. In addition, NK-EVs and NK-EVsIL-15 were characterized by transmission electron microscopy, nanoparticle-tracking analysis, and western blotting. Flow cytometry, bioluminescence imaging, and a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay were performed for apoptosis, protein expression, cell proliferation, and cytotoxicity analyses. Furthermore, xenograft tumor-bearing mice were injected with PBS, NK-EVs, or NK-EVsIL-15 intravenously five times. Tumor growth was monitored using calipers and bioluminescence imaging. Toxicity of the nanoparticles was evaluated by measuring the body weight of the mice. RESULTS NK-EVsIL-15 showed significantly higher cytolytic activity toward human cancer cell lines (glioblastoma, breast cancer, and thyroid cancer) and simultaneously increased the expression of molecules associated with NK-cell cytotoxicity. When compared with NK-EVs, NK-EVsIL-15 significantly inhibited the growth of glioblastoma xenograft cells in mice. In addition, both NK-EVs and NK-EVsIL-15 were not significantly toxic to either normal cells or mice. CONCLUSION IL-15 may improve the immunotherapeutic effects of NK-EVs, thus improving the applications of NK-EVs in the future.
Collapse
Affiliation(s)
- Liya Zhu
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, South Korea
| | - Senthilkumar Kalimuthu
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, South Korea
| | - Ji Min Oh
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, South Korea
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, South Korea
| | - Se Hwan Baek
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, South Korea
| | - Shin Young Jeong
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, South Korea
| | - Sang-Woo Lee
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, South Korea
| | - Jaetae Lee
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, South Korea
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, South Korea.
| |
Collapse
|
32
|
Rautela J, Souza-Fonseca-Guimaraes F, Hediyeh-Zadeh S, Delconte RB, Davis MJ, Huntington ND. Molecular insight into targeting the NK cell immune response to cancer. Immunol Cell Biol 2018; 96:477-484. [PMID: 29577414 DOI: 10.1111/imcb.12045] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 03/17/2018] [Accepted: 03/18/2018] [Indexed: 02/06/2023]
Abstract
Natural Killer (NK) cells have long been considered an important part of the anti-tumor immune response due to their potent cytolytic and cytokine-secreting abilities. To date, a clear demonstration of the role NK cells play in human cancer is lacking, and there are still very few examples of therapies that efficiently exploit or enhance the spontaneous ability of NK cells to destroy the autologous cancer cells. Given the paradigm shift toward cancer immunotherapy over the past decade, there is a renewed push to understand how NK cell homeostasis and function are regulated in order to therapeutically harness these cells to treat cancer. This review will highlight recent advancements in our understanding of how growth factors impact on NK cell development, differentiation, survival and function with an emphasis on how these pathways may influence NK cell activity in the tumor microenvironment and control of cancer metastasis.
Collapse
Affiliation(s)
- Jai Rautela
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Fernando Souza-Fonseca-Guimaraes
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Soroor Hediyeh-Zadeh
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Rebecca B Delconte
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Melissa J Davis
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Nicholas D Huntington
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
33
|
Schmohl JU, Felices M, Todhunter D, Taras E, Miller JS, Vallera DA. Tetraspecific scFv construct provides NK cell mediated ADCC and self-sustaining stimuli via insertion of IL-15 as a cross-linker. Oncotarget 2018; 7:73830-73844. [PMID: 27650544 PMCID: PMC5342017 DOI: 10.18632/oncotarget.12073] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 09/05/2016] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND The design of a highly effective anti-cancer immune-engager would include targeting of highly drug refractory cancer stem cells (CSC). The design would promote effective antibody-dependent cell-mediated cytotoxicity (ADCC) and simultaneously promote costimulation to expand and self-sustain the effector NK cell population. Based on our bispecific NK cell engager platform we constructed a tetraspecific killer engager (TetraKE) comprising single-chain variable fragments (scFvs) binding FcγRIII (CD16) on NK cells, EpCAM on carcinoma cells and CD133 on cancer stem cells in order to promote ADCC. Furthermore, an Interleukin (IL)-15-crosslinker enhanced NK cell related proliferation resulting in a highly active drug termed 1615EpCAM133. RESULTS Proliferation assays showed TetraKE promoted proliferation and enhanced NK cell survival. Drug-target binding, NK cell related degranulation, and IFN-γ production was specific for both tumor related antigens in EpCAM and CD133 bearing cancer cell lines. The TetraKE showed higher killing activity and superior dose dependent degranulation. Cytokine profiling showed a moderately enhanced IFN-γ production, enhanced GM-CSF production, but no evidence of induction of excessive cytokine release. METHODS Assembly and synthesis of hybrid genes encoding the TetraKE were performed using DNA shuffling and ligation. The TetraKE was tested for efficacy, specificity, proliferation, survival, and cytokine production using carcinoma cell lines and functional assays measuring NK cell activity. CONCLUSION 1615EpCAM133 combines improved induction of ADCC with enhanced proliferation, limited cytokine response, and prolonged survival and proliferation of NK cells. By linking scFv-related targeting of carcinoma and CSCs with a sustaining IL-15 signal, our new construct shows great promise to target cancer and CSCs.
Collapse
Affiliation(s)
- Joerg U Schmohl
- University of Minnesota, Masonic Cancer Center, Section of Molecular Cancer Therapeutics, Therapeutic Radiology-Radiation Oncology, University of Minnesota, Minneapolis, MN, USA.,University of Tuebingen, Department for Hematology and Oncology, Medicine Department 2, University Hospital of Tuebingen, Tuebingen, Germany
| | - Martin Felices
- University of Minnesota, Department of Medicine, Division of Hematology, Oncology, and Transplantation, Minneapolis, MN, USA
| | - Deborah Todhunter
- University of Minnesota, Masonic Cancer Center, Section of Molecular Cancer Therapeutics, Therapeutic Radiology-Radiation Oncology, University of Minnesota, Minneapolis, MN, USA
| | - Elizabeth Taras
- University of Minnesota, Masonic Cancer Center, Section of Molecular Cancer Therapeutics, Therapeutic Radiology-Radiation Oncology, University of Minnesota, Minneapolis, MN, USA
| | - Jeffrey S Miller
- University of Minnesota, Department of Medicine, Division of Hematology, Oncology, and Transplantation, Minneapolis, MN, USA
| | - Daniel A Vallera
- University of Minnesota, Masonic Cancer Center, Section of Molecular Cancer Therapeutics, Therapeutic Radiology-Radiation Oncology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
34
|
Wrangle JM, Patterson A, Johnson CB, Neitzke DJ, Mehrotra S, Denlinger CE, Paulos CM, Li Z, Cole DJ, Rubinstein MP. IL-2 and Beyond in Cancer Immunotherapy. J Interferon Cytokine Res 2018; 38:45-68. [PMID: 29443657 PMCID: PMC5815463 DOI: 10.1089/jir.2017.0101] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 11/26/2017] [Indexed: 12/11/2022] Open
Abstract
The development of the T- and natural killer (NK) cell growth factor IL-2 has been a sentinel force ushering in the era of immunotherapy in cancer. With the advent of clinical grade recombinant IL-2 in the mid-1980s, oncologists could for the first time directly manipulate lymphocyte populations with systemic therapy. By itself, recombinant IL-2 can induce clinical responses in up to 15% of patients with metastatic cancer or renal cell carcinoma. When administered with adoptively transferred tumor-reactive lymphocytes, IL-2 promotes T cell engraftment and response rates of up to 50% in metastatic melanoma patients. Importantly, these IL-2-driven responses can yield complete and durable responses in a subset of patients. However, the use of IL-2 is limited by toxicity and concern of the expansion of T regulatory cells. To overcome these limitations and improve response rates, other T cell growth factors, including IL-15 and modified forms of IL-2, are in clinical development. Administering T cell growth factors in combination with other agents, such as immune checkpoint pathway inhibitors, may also improve efficacy. In this study, we review the development of T- and NK cell growth factors and highlight current combinatorial approaches based on these reagents.
Collapse
Affiliation(s)
- John M. Wrangle
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Alicia Patterson
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - C. Bryce Johnson
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Daniel J. Neitzke
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Shikhar Mehrotra
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Chadrick E. Denlinger
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Chrystal M. Paulos
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Zihai Li
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - David J. Cole
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Mark P. Rubinstein
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
35
|
Wang Z, Han W. Biomarkers of cytokine release syndrome and neurotoxicity related to CAR-T cell therapy. Biomark Res 2018; 6:4. [PMID: 29387417 PMCID: PMC5778792 DOI: 10.1186/s40364-018-0116-0] [Citation(s) in RCA: 186] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 01/02/2018] [Indexed: 12/22/2022] Open
Abstract
Severe cytokine release syndrome (CRS) and neurotoxicity following chimeric antigen receptor T cell (CAR-T) therapy can be life-threatening in some cases, and management of those toxicities is still a great challenge for physicians. Researchers hope to understand the pathophysiology of CRS and neurotoxicity, and identify predictive biomarkers that can forecast those toxicities in advance. Some risk factors for severe CRS and/or neurotoxicity including patient and treatment characteristics have been identified in multiple clinical trials of CAR-T cell therapy. Moreover, several groups have identified some predictive biomarkers that are able to determine beforehand which patients may suffer severe CRS and/or neurotoxicity during CAR-T cell therapy, facilitating testing of early intervention strategies for those toxicities. However, further studies are needed to better understand the biology and related risk factors for CRS and/or neurotoxicity, and determine if those identified predictors can be extrapolated to other series. Herein, we review the pathophysiology of CRS and neurotoxicity, and summarize the progress of predictive biomarkers to improve CAR-T cell therapy in cancer.
Collapse
Affiliation(s)
- Zhenguang Wang
- Molecular & Immunological Department, Bio-therapeutic Department, Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853 China
| | - Weidong Han
- Molecular & Immunological Department, Bio-therapeutic Department, Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853 China
| |
Collapse
|
36
|
Fercher C, Keshvari S, McGuckin MA, Barnard RT. Evolution of the magic bullet: Single chain antibody fragments for the targeted delivery of immunomodulatory proteins. Exp Biol Med (Maywood) 2017; 243:166-183. [PMID: 29256259 DOI: 10.1177/1535370217748575] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Immunocytokines are fusion proteins that combine the specific antigen binding capacities of an antibody or derivative thereof and the potent bioactivity of a cytokine partner. These novel biopharmaceuticals have been directed to various targets of oncological as well as non-oncological origin and a handful of promising constructs are currently advancing in the clinical trial pipeline. Several factors such as the choice of a disease specific antigen, the antibody format and the modulatory nature of the payload are crucial, not only for therapeutic efficacy and safety but also for the commercial success of such a product. In this review, we provide an overview of the basic principles and obstacles in immunocytokine design with a specific focus on single chain antibody fragment-based constructs that employ interleukins as the immunoactive component. Impact statement Selective activation of the immune system in a variety of malignancies represents an attractive approach when existing strategies have failed to provide adequate treatment options. Immunocytokines as a novel class of bifunctional protein therapeutics have emerged recently and generated promising results in preclinical and clinical studies. In order to harness their full potential, multiple different aspects have to be taken into consideration. Several key points of these fusion constructs are discussed here and should provide an outline for the development of novel products based on an overview of selected formats.
Collapse
Affiliation(s)
- Christian Fercher
- 1 School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Sahar Keshvari
- 2 Inflammatory Diseases Biology and Therapeutics, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Michael A McGuckin
- 2 Inflammatory Diseases Biology and Therapeutics, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Ross T Barnard
- 1 School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia.,3 Australian Research Council Training Centre for Biopharmaceutical Innovation, The University of Queensland, St. Lucia, QLD 4072, Australia
| |
Collapse
|
37
|
Rao M, Zhenjiang L, Meng Q, Sinclair G, Dodoo E, Maeurer M. Mutant Epitopes in Cancer. Oncoimmunology 2017. [DOI: 10.1007/978-3-319-62431-0_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
38
|
Association between Recipient IL-15 Genetic Variant and the Prognosis of HBV-Related Hepatocellular Carcinoma after Liver Transplantation. DISEASE MARKERS 2017; 2017:1754696. [PMID: 29162948 PMCID: PMC5661074 DOI: 10.1155/2017/1754696] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 08/20/2017] [Accepted: 09/14/2017] [Indexed: 02/06/2023]
Abstract
Objective To investigate the association of donor and recipient IL-15 genetic variants with HCC recurrence and prognosis after LT. Methods A total of 112 liver transplant patients with HBV-related HCC were enrolled. IL-15 rs10519613 and rs13122930 were genotyped in donors and recipients. Results Recipient IL-15 rs10519613 polymorphism was found to be significantly related to HCC recurrence after LT. In multivariate analysis, tumor thrombus, UCSF criteria, and recipient IL-15 rs10519613 genotypes were independent predictive factors of HCC recurrence after LT. Kaplan-Meier survival analysis demonstrated that patients with recipient IL-15 rs10519613 CA/AA genotypes had a decreased disease-free survival and overall survival than those with the CC genotype. Recipient IL-15 rs10519613 genetic variant could improve survival prediction when combined with the UCSF criteria. Furthermore, Cox proportional hazard regression analysis revealed that tumor size (p = 0.012, p = 0.623), tumor thrombus (p = 0.011, p = 0.015), UCSF criteria (p = 0.471, p = 0.013), and recipient IL-15 rs10519613 genotype (p = 0.039, p = 0.008) were independent factors of predicting DFS and OS. Conclusions Recipient IL-15 rs10519613 polymorphism was associated with HCC recurrence after LT and might be a potential genetic marker for the clinical outcome of HCC patients treated with LT.
Collapse
|
39
|
Wagner JA, Rosario M, Romee R, Berrien-Elliott MM, Schneider SE, Leong JW, Sullivan RP, Jewell BA, Becker-Hapak M, Schappe T, Abdel-Latif S, Ireland AR, Jaishankar D, King JA, Vij R, Clement D, Goodridge J, Malmberg KJ, Wong HC, Fehniger TA. CD56bright NK cells exhibit potent antitumor responses following IL-15 priming. J Clin Invest 2017; 127:4042-4058. [PMID: 28972539 DOI: 10.1172/jci90387] [Citation(s) in RCA: 201] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 08/15/2017] [Indexed: 12/12/2022] Open
Abstract
NK cells, lymphocytes of the innate immune system, are important for defense against infectious pathogens and cancer. Classically, the CD56dim NK cell subset is thought to mediate antitumor responses, whereas the CD56bright subset is involved in immunomodulation. Here, we challenge this paradigm by demonstrating that brief priming with IL-15 markedly enhanced the antitumor response of CD56bright NK cells. Priming improved multiple CD56bright cell functions: degranulation, cytotoxicity, and cytokine production. Primed CD56bright cells from leukemia patients demonstrated enhanced responses to autologous blasts in vitro, and primed CD56bright cells controlled leukemia cells in vivo in a murine xenograft model. Primed CD56bright cells from multiple myeloma (MM) patients displayed superior responses to autologous myeloma targets, and furthermore, CD56bright NK cells from MM patients primed with the IL-15 receptor agonist ALT-803 in vivo displayed enhanced ex vivo functional responses to MM targets. Effector mechanisms contributing to IL-15-based priming included improved cytotoxic protein expression, target cell conjugation, and LFA-1-, CD2-, and NKG2D-dependent activation of NK cells. Finally, IL-15 robustly stimulated the PI3K/Akt/mTOR and MEK/ERK pathways in CD56bright compared with CD56dim NK cells, and blockade of these pathways attenuated antitumor responses. These findings identify CD56bright NK cells as potent antitumor effectors that warrant further investigation as a cancer immunotherapy.
Collapse
Affiliation(s)
- Julia A Wagner
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Maximillian Rosario
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Rizwan Romee
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Melissa M Berrien-Elliott
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Stephanie E Schneider
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jeffrey W Leong
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ryan P Sullivan
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Brea A Jewell
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Michelle Becker-Hapak
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Timothy Schappe
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sara Abdel-Latif
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Aaron R Ireland
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Devika Jaishankar
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Justin A King
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ravi Vij
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Dennis Clement
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Radiumhospitalet, Oslo, Norway.,The KG Jebsen Centre for Cancer Immunotherapy, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Jodie Goodridge
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Radiumhospitalet, Oslo, Norway
| | - Karl-Johan Malmberg
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Radiumhospitalet, Oslo, Norway.,The KG Jebsen Centre for Cancer Immunotherapy, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Centre for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | | | - Todd A Fehniger
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
40
|
Bozorg-Ghalati F, Hedayati M, Dianatpour M, Azizi F, Mosaffa N, Mehrabani D. Effects of a Phosphoinositide-3-Kinase Inhibitor on Anaplastic Thyroid Cancer Stem Cells. Asian Pac J Cancer Prev 2017; 18:2287-2291. [PMID: 28843268 PMCID: PMC5697493 DOI: 10.22034/apjcp.2017.18.8.2287] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Background: Thyroidectomy, radioactive iodine therapy, chemotherapy, or their combination are treatments of choice for thyroid cancers. However, cancer stem cells (CSCs) may become resistant to therapy, and mutations in somatic genes affect radioiodine uptake. This study determined the effect of a phosphoinositide-3-kinase (PI3K) inhibitor on anaplastic thyroid CSCs. Materials and Methods: The magnetic-activated cell sorting assay was used for segregating CD133-positive CSCs from three anaplastic thyroid carcinoma (ATC) cell lines (C643, SW1736, and 8305C). After confirming the cells’ purity by flow cytometry, they were treated with 5, 10, 20, or 25 μM LY294002, a PI3K inhibitor, and then evaluated at 24 and 48 h. The sodium-iodide symporter (NIS) mRNA level was determined using the quantitative real-time polymerase chain reaction. NIS protein expression was evaluated using western blotting. Results: The PI3K inhibitor, at different concentrations and times, increased the NIS mRNA level (1.30-6.17-fold, P < 0.0001). If the NIS mRNA level in LY294002-treated CD133-positive CSCs was increased more than 2-fold, the NIS protein content was detectable. Conclusions: CD133-positive CSCs isolated from ATC cell lines expressed NIS mRNA and protein after PI3K inhibition. Our findings suggest that molecularly targeted CSC therapy may improve the treatment efficacy of aggressive cancers like ATC.
Collapse
Affiliation(s)
- Farzaneh Bozorg-Ghalati
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran. , dianatpour@sums.
ac.ir
| | | | | | | | | | | |
Collapse
|
41
|
Van Acker HH, Capsomidis A, Smits EL, Van Tendeloo VF. CD56 in the Immune System: More Than a Marker for Cytotoxicity? Front Immunol 2017; 8:892. [PMID: 28791027 PMCID: PMC5522883 DOI: 10.3389/fimmu.2017.00892] [Citation(s) in RCA: 222] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Accepted: 07/12/2017] [Indexed: 11/13/2022] Open
Abstract
Over the past years, the phenotypic and functional boundaries distinguishing the main cell subsets of the immune system have become increasingly blurred. In this respect, CD56 (also known as neural cell adhesion molecule) is a very good example. CD56 is the archetypal phenotypic marker of natural killer cells but can actually be expressed by many more immune cells, including alpha beta T cells, gamma delta T cells, dendritic cells, and monocytes. Common to all these CD56-expressing cell types are strong immunostimulatory effector functions, including T helper 1 cytokine production and an efficient cytotoxic capacity. Interestingly, both numerical and functional deficiencies and phenotypic alterations of the CD56+ immune cell fraction have been reported in patients with various infectious, autoimmune, or malignant diseases. In this review, we will discuss our current knowledge on the expression and function of CD56 in the hematopoietic system, both in health and disease.
Collapse
Affiliation(s)
- Heleen H Van Acker
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| | - Anna Capsomidis
- Cancer Section, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Evelien L Smits
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium.,Center for Oncological Research (CORE), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Viggo F Van Tendeloo
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| |
Collapse
|
42
|
Guillerey C, Huntington ND, Smyth MJ. Targeting natural killer cells in cancer immunotherapy. Nat Immunol 2017; 17:1025-36. [PMID: 27540992 DOI: 10.1038/ni.3518] [Citation(s) in RCA: 767] [Impact Index Per Article: 109.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 06/22/2016] [Indexed: 12/14/2022]
Abstract
Alteration in the expression of cell-surface proteins is a common consequence of malignant transformation. Natural killer (NK) cells use an array of germline-encoded activating and inhibitory receptors that scan for altered protein-expression patterns, but tumor evasion of detection by the immune system is now recognized as one of the hallmarks of cancer. NK cells display rapid and potent immunity to metastasis or hematological cancers, and major efforts are now being undertaken to fully exploit NK cell anti-tumor properties in the clinic. Diverse approaches encompass the development of large-scale NK cell-expansion protocols for adoptive transfer, the establishment of a microenvironment favorable to NK cell activity, the redirection of NK cell activity against tumor cells and the release of inhibitory signals that limit NK cell function. In this Review we detail recent advances in NK cell-based immunotherapies and discuss the advantages and limitations of these strategies.
Collapse
Affiliation(s)
- Camille Guillerey
- Immunology of Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Australia.,School of Medicine, University of Queensland, Herston, Australia
| | - Nicholas D Huntington
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, Australia
| | - Mark J Smyth
- Immunology of Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Australia.,School of Medicine, University of Queensland, Herston, Australia
| |
Collapse
|
43
|
Rohena-Rivera K, Sánchez-Vázquez MM, Aponte-Colón DA, Forestier-Román IS, Quintero-Aguiló ME, Martínez-Ferrer M. IL-15 regulates migration, invasion, angiogenesis and genes associated with lipid metabolism and inflammation in prostate cancer. PLoS One 2017; 12:e0172786. [PMID: 28379958 PMCID: PMC5381801 DOI: 10.1371/journal.pone.0172786] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Accepted: 02/09/2017] [Indexed: 01/16/2023] Open
Abstract
Prostate cancer (PCa) is the most commonly diagnosed non-cutaneous cancer. In the United States it is second leading cause of cancer related deaths in men. PCa is often treated via radical prostatectomy (RP). However, 15–30% of the patients develop biochemical recurrence (i.e. increased serum prostate specific antigen (PSA) levels). Interleukin-15 (IL-15) is a secreted cytokine found over expressed in patients with recurrence-free survival after RP. In our study, we aim to determine the role of IL-15 in PCa using in vitro and in vivo models, and gene expression analysis. PC3 (androgen-independent) and 22RV1 (androgen-dependent) cell lines were treated with IL-15 at 0.0013 ng/mL and 0.1 ng/mL. Tumor growth was evaluated using an orthotopic xenograft model. The anterior prostate lobes of SCID mice were injected with 250,000 22RV1 cells and IL-15 was administered bi-weekly with intraperitoneal (IP) injections during 4 weeks. Tumor tissue was collected for immunohistochemical and gene expression analysis. To study changes in gene expression, we looked at “Tumor Metastasis” and “PI3K pathway” using commercially available PCR arrays. In addition, we employed a microarray approach using the Affymetrix Hugene 2.0 ST array chip followed by analysis with Ingenuity Pathways Analysis (IPA) software. In vitro studies showed that IL-15 decreased PCa cell motility at both concentrations. In vivo studies showed that IL-15 increased neutrophil infiltration, and the expression of adiponectin, desmin and alpha smooth muscle actin (α-sma) in the tumor tissue. Angiogenesis analysis, using CD31 immunohistochemistry, showed that IL-15 decreased the number of blood vessels. Gene expression analysis identified Cancer, Cell Death, Immune Response and Lipid Metabolism as the major diseases and functions altered in tumors treated with IL-15. This suggests that IL-15 causes inflammation and changes in stroma that can promote decreased tumor cell proliferation.
Collapse
Affiliation(s)
- Krizia Rohena-Rivera
- Department of Biochemistry, University of Puerto Rico, School of Medicine, San Juan, Puerto Rico
- University of Puerto Rico Comprehensive Cancer Center San Juan, Puerto Rico
| | | | | | - Ingrid S. Forestier-Román
- Department of Biochemistry, University of Puerto Rico, School of Medicine, San Juan, Puerto Rico
- University of Puerto Rico Comprehensive Cancer Center San Juan, Puerto Rico
| | | | - Magaly Martínez-Ferrer
- Department of Biochemistry, University of Puerto Rico, School of Medicine, San Juan, Puerto Rico
- University of Puerto Rico Comprehensive Cancer Center San Juan, Puerto Rico
- Department of Pharmaceutical Sciences, University of Puerto Rico, School of Pharmacy San Juan, Puerto Rico
- * E-mail:
| |
Collapse
|
44
|
Kochenderfer JN, Somerville RPT, Lu T, Shi V, Bot A, Rossi J, Xue A, Goff SL, Yang JC, Sherry RM, Klebanoff CA, Kammula US, Sherman M, Perez A, Yuan CM, Feldman T, Friedberg JW, Roschewski MJ, Feldman SA, McIntyre L, Toomey MA, Rosenberg SA. Lymphoma Remissions Caused by Anti-CD19 Chimeric Antigen Receptor T Cells Are Associated With High Serum Interleukin-15 Levels. J Clin Oncol 2017; 35:1803-1813. [PMID: 28291388 DOI: 10.1200/jco.2016.71.3024] [Citation(s) in RCA: 422] [Impact Index Per Article: 60.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Purpose T cells genetically modified to express chimeric antigen receptors (CARs) targeting CD19 (CAR-19) have potent activity against acute lymphoblastic leukemia, but fewer results supporting treatment of lymphoma with CAR-19 T cells have been published. Patients with lymphoma that is chemotherapy refractory or relapsed after autologous stem-cell transplantation have a grim prognosis, and new treatments for these patients are clearly needed. Chemotherapy administered before adoptive T-cell transfer has been shown to enhance the antimalignancy activity of adoptively transferred T cells. Patients and Methods We treated 22 patients with advanced-stage lymphoma in a clinical trial of CAR-19 T cells preceded by low-dose chemotherapy. Nineteen patients had diffuse large B-cell lymphoma, two patients had follicular lymphoma, and one patient had mantle cell lymphoma. Patients received a single dose of CAR-19 T cells 2 days after a low-dose chemotherapy conditioning regimen of cyclophosphamide plus fludarabine. Results The overall remission rate was 73% with 55% complete remissions and 18% partial remissions. Eleven of 12 complete remissions are ongoing. Fifty-five percent of patients had grade 3 or 4 neurologic toxicities that completely resolved. The low-dose chemotherapy conditioning regimen depleted blood lymphocytes and increased serum interleukin-15 (IL-15). Patients who achieved a remission had a median peak blood CAR+ cell level of 98/μL and those who did not achieve a remission had a median peak blood CAR+ cell level of 15/μL ( P = .027). High serum IL-15 levels were associated with high peak blood CAR+ cell levels ( P = .001) and remissions of lymphoma ( P < .001). Conclusion CAR-19 T cells preceded by low-dose chemotherapy induced remission of advanced-stage lymphoma, and high serum IL-15 levels were associated with the effectiveness of this treatment regimen. CAR-19 T cells will likely become an important treatment for patients with relapsed lymphoma.
Collapse
Affiliation(s)
- James N Kochenderfer
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Robert P T Somerville
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Tangying Lu
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Victoria Shi
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Adrian Bot
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - John Rossi
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Allen Xue
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Stephanie L Goff
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - James C Yang
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Richard M Sherry
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Christopher A Klebanoff
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Udai S Kammula
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Marika Sherman
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Arianne Perez
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Constance M Yuan
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Tatyana Feldman
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Jonathan W Friedberg
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Mark J Roschewski
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Steven A Feldman
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Lori McIntyre
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Mary Ann Toomey
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Steven A Rosenberg
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| |
Collapse
|
45
|
Donahue RN, Lepone LM, Grenga I, Jochems C, Fantini M, Madan RA, Heery CR, Gulley JL, Schlom J. Analyses of the peripheral immunome following multiple administrations of avelumab, a human IgG1 anti-PD-L1 monoclonal antibody. J Immunother Cancer 2017; 5:20. [PMID: 28239472 PMCID: PMC5320726 DOI: 10.1186/s40425-017-0220-y] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 02/01/2017] [Indexed: 12/11/2022] Open
Abstract
Background Multiple anti-PD-L1/PD-1 checkpoint monoclonal antibodies (MAb) have shown clear evidence of clinical benefit. All except one have been designed or engineered to omit the possibility to mediate antibody-dependent cell-mediated cytotoxicity (ADCC) as a second potential mode of anti-tumor activity; the reason for this is the concern of lysis of PD-L1 positive immune cells. Avelumab is a fully human IgG1 MAb which has been shown in prior in vitro studies to mediate ADCC versus a range of human tumor cells, and clinical studies have demonstrated anti-tumor activity versus a range of human cancers. This study was designed to investigate the effect on immune cell subsets in the peripheral blood of cancer patients prior to and following multiple administrations of avelumab. Methods One hundred twenty-three distinct immune cell subsets in the peripheral blood of cancer patients (n = 28) in a phase I trial were analyzed by flow cytometry prior to and following one, three, and nine cycles of avelumab. Changes in soluble (s) CD27 and sCD40L in plasma were also evaluated. In vitro studies were also performed to determine if avelumab would mediate ADCC of PBMC. Results No statistically significant changes in any of the 123 immune cell subsets analyzed were observed at any dose level, or number of doses, of avelumab. Increases in the ratio of sCD27:sCD40L were observed, suggesting potential immune activation. Controlled in vitro studies also showed lysis of tumor cells by avelumab versus no lysis of PBMC from five donors. Conclusions These studies demonstrate the lack of any significant effect on multiple immune cell subsets, even those expressing PD-L1, following multiple cycles of avelumab. These results complement prior studies showing anti-tumor effects of avelumab and comparable levels of adverse events with avelumab versus other anti-PD-1/PD-L1 MAbs. These studies provide the rationale to further exploit the potential ADCC mechanism of action of avelumab as well as other human IgG1 checkpoint inhibitors. Trial registration ClinicalTrials.gov identifier: NCT01772004 (first received: 1/14/13; start date: January 2013) and NCT00001846 (first received date: 11/3/99; start date: August 1999). Electronic supplementary material The online version of this article (doi:10.1186/s40425-017-0220-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Renee N Donahue
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B09, Bethesda, MD USA
| | - Lauren M Lepone
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B09, Bethesda, MD USA
| | - Italia Grenga
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B09, Bethesda, MD USA
| | - Caroline Jochems
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B09, Bethesda, MD USA
| | - Massimo Fantini
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B09, Bethesda, MD USA
| | - Ravi A Madan
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD USA
| | - Christopher R Heery
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B09, Bethesda, MD USA
| | - James L Gulley
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD USA
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B09, Bethesda, MD USA
| |
Collapse
|
46
|
Schmohl JU, Felices M, Oh F, Lenvik AJ, Lebeau AM, Panyam J, Miller JS, Vallera DA. Engineering of Anti-CD133 Trispecific Molecule Capable of Inducing NK Expansion and Driving Antibody-Dependent Cell-Mediated Cytotoxicity. Cancer Res Treat 2017; 49:1140-1152. [PMID: 28231426 PMCID: PMC5654165 DOI: 10.4143/crt.2016.491] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 01/30/2017] [Indexed: 01/19/2023] Open
Abstract
Purpose The selective elimination of cancer stem cells (CSCs) in tumor patients is a crucial goal because CSCs cause drug refractory relapse. To improve the current conventional bispecific immune-engager platform, a 16133 bispecific natural killer (NK) cell engager (BiKE), consisting of scFvs binding FcγRIII (CD16) on NK cells and CD133 on carcinoma cells, was first synthesized and a modified interleukin (IL)-15 crosslinker capable of stimulating NK effector cells was introduced. Materials and Methods DNA shuffling and ligation techniques were used to assemble and synthesize the 1615133 trispecific NK cell engager (TriKE). The construct was tested for its specificity using flow cytometry, cytotoxic determinations using chromium release assays, and lytic degranulation. IL-15–mediated expansion was measured using flow-based proliferation assays. The level of interferon (IFN)-γ release was measured because of its importance in the anti-cancer response. Results 1615133 TriKE induced NK cell–mediated cytotoxicity and NK expansion far greater than that achieved with BiKE devoid of IL-15. The drug binding and induction of cytotoxic degranulation was CD133+ specific and the anti-cancer activity was improved by integrating the IL-15 cross linker. The NK cell–related cytokine release measured by IFN-γ detection was higher than that of BiKE. NK cytokine release studies showed that although the IFN-γ levels were elevated, they did not approach the levels achieved with IL-12/IL-18, indicating that release was not at the supraphysiologic level. Conclusion 1615133 TriKE enhances the NK cell anti-cancer activity and provides a self-sustaining mechanism via IL-15 signaling. By improving the NK cell performance, the new TriKE represents a highly active drug against drug refractory relapse mediated by CSCs.
Collapse
Affiliation(s)
- Jörg U Schmohl
- Department of Therapeutic Radiology-Radiation Oncology, University of Minnesota, Masonic Cancer Center, Minneapolis, MN, USA.,Department for Hematology and Oncology, Medicine Department 2, University Hospital of Tuebingen, University of Tuebingen, Tuebingen, Germany
| | - Martin Felices
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, Minneapolis, MN, USA
| | - Felix Oh
- Department of Therapeutic Radiology-Radiation Oncology, University of Minnesota, Masonic Cancer Center, Minneapolis, MN, USA
| | - Alexander J Lenvik
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, Minneapolis, MN, USA
| | - Aaron M Lebeau
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - Jayanth Panyam
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - Jeffrey S Miller
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, Minneapolis, MN, USA
| | - Daniel A Vallera
- Department of Therapeutic Radiology-Radiation Oncology, University of Minnesota, Masonic Cancer Center, Minneapolis, MN, USA
| |
Collapse
|
47
|
Tallerico R, Cristiani CM, Staaf E, Garofalo C, Sottile R, Capone M, Pico de Coaña Y, Madonna G, Palella E, Wolodarski M, Carannante V, Mallardo D, Simeone E, Grimaldi AM, Johansson S, Frumento P, Gulletta E, Anichini A, Colucci F, Ciliberto G, Kiessling R, Kärre K, Ascierto PA, Carbone E. IL-15, TIM-3 and NK cells subsets predict responsiveness to anti-CTLA-4 treatment in melanoma patients. Oncoimmunology 2016; 6:e1261242. [PMID: 28344869 DOI: 10.1080/2162402x.2016.1261242] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 11/09/2016] [Accepted: 11/09/2016] [Indexed: 12/31/2022] Open
Abstract
Despite the success of immune checkpoint blockade in melanoma, the majority of patients do not respond. We hypothesized that the T and NK cell subset frequencies and expression levels of their receptors may predict responses and clinical outcome of anti-CTLA-4 treatment. We thus characterized the NK and T cell phenotype, as well as serum levels of several cytokines in 67 melanoma patients recruited in Italy and Sweden, using samples drawn prior to and during treatment. Survival correlated with low expression of the inhibitory receptor TIM-3 on circulating T and NK cells prior to and during treatment and with the increased frequency of mature circulating NK cells (defined as CD3-CD56dim CD16+) during treatment. Survival also correlated with low levels of IL-15 in the serum. Functional experiments in vitro demonstrated that sustained exposure to IL-15 enhanced the expression of PD-1 and TIM-3 on both T and NK cells, indicating a causative link between high IL-15 levels and enhanced expression of TIM-3 on these cells. Receptor blockade of TIM-3 improved NK cell-mediated elimination of melanoma metastasis cell lines in vitro. These observations may lead to the development of novel biomarkers to predict patient response to checkpoint blockade treatment. They also suggest that induction of additional checkpoints is a possibility that needs to be considered when treating melanoma patients with IL-15.
Collapse
Affiliation(s)
- Rossana Tallerico
- Tumor Immunology and Immunopathology Laboratory, Department of Experimental and Clinical Medicine, University "Magna Græcia" of Catanzaro, Campus - Germaneto , Catanzaro, Italy
| | - Costanza M Cristiani
- Tumor Immunology and Immunopathology Laboratory, Department of Experimental and Clinical Medicine, University "Magna Græcia" of Catanzaro, Campus - Germaneto , Catanzaro, Italy
| | - Elina Staaf
- Department of Microbiology, Cell and Tumorbiology (MTC), Karolinska Institutet , Stockholm, Sweden
| | - Cinzia Garofalo
- Tumor Immunology and Immunopathology Laboratory, Department of Experimental and Clinical Medicine, University "Magna Græcia" of Catanzaro, Campus - Germaneto , Catanzaro, Italy
| | - Rosa Sottile
- Tumor Immunology and Immunopathology Laboratory, Department of Experimental and Clinical Medicine, University "Magna Græcia" of Catanzaro, Campus - Germaneto, Catanzaro, Italy; Department of Microbiology, Cell and Tumorbiology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Mariaelena Capone
- Melanoma Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori Fondazione "G. Pascale ," Napoli, Italy
| | - Yago Pico de Coaña
- Department of Oncology and Pathology, Karolinska Institutet , Stockholm, Sweden
| | - Gabriele Madonna
- Melanoma Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori Fondazione "G. Pascale ," Napoli, Italy
| | - Eleonora Palella
- Tumor Immunology and Immunopathology Laboratory, Department of Experimental and Clinical Medicine, University "Magna Græcia" of Catanzaro, Campus - Germaneto , Catanzaro, Italy
| | - Maria Wolodarski
- Department of Oncology and Pathology, Karolinska Institutet , Stockholm, Sweden
| | - Valentina Carannante
- Department of Microbiology, Cell and Tumorbiology (MTC), Karolinska Institutet , Stockholm, Sweden
| | - Domenico Mallardo
- Melanoma Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori Fondazione "G. Pascale ," Napoli, Italy
| | - Ester Simeone
- Melanoma Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori Fondazione "G. Pascale ," Napoli, Italy
| | - Antonio M Grimaldi
- Melanoma Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori Fondazione "G. Pascale ," Napoli, Italy
| | - Sofia Johansson
- Department of Microbiology, Cell and Tumorbiology (MTC), Karolinska Institutet , Stockholm, Sweden
| | - Paolo Frumento
- Karolinska Institutet Statistical Core Facility, Karolinska Institutet , Stockholm, Sweden
| | - Elio Gulletta
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Campus - Germaneto , Catanzaro, Italy
| | - Andrea Anichini
- Fondazione IRCCS Istituto Nazionale dei Tumori, Department of Experimental Oncology and Molecular Medicine , Milan, Italy
| | - Francesco Colucci
- Department of Obstetrics and Gynecology, University of Cambridge Clinical School , Cambridge, UK
| | - Gennaro Ciliberto
- Scientific Directorate, IRCCS Istituto Nazionale Tumori Fondazione "G. Pascale ," Napoli, Italy
| | - Rolf Kiessling
- Department of Oncology and Pathology, Karolinska Institutet , Stockholm, Sweden
| | - Klas Kärre
- Department of Microbiology, Cell and Tumorbiology (MTC), Karolinska Institutet , Stockholm, Sweden
| | - Paolo A Ascierto
- Melanoma Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori Fondazione "G. Pascale ," Napoli, Italy
| | - Ennio Carbone
- Tumor Immunology and Immunopathology Laboratory, Department of Experimental and Clinical Medicine, University "Magna Græcia" of Catanzaro, Campus - Germaneto, Catanzaro, Italy; Department of Microbiology, Cell and Tumorbiology (MTC), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
48
|
Rautela J, Huntington ND. IL-15 signaling in NK cell cancer immunotherapy. Curr Opin Immunol 2016; 44:1-6. [PMID: 27835762 DOI: 10.1016/j.coi.2016.10.004] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 10/28/2016] [Indexed: 10/20/2022]
Abstract
While cancer has been traditionally treated by chemotherapy, radiation, targeted therapies and surgery, a fifth pillar of cancer treatment, immunotherapy, has emerged over the past 10 years and revolutionized our war on cancer. The benchmark for drugs in this category has been set by the development of CD8 T cell checkpoint (CTLA-4 and PD-1/PD-L1) inhibitors. These therapies effectively expand and reactivate the pool of tumor-specific T cells leading to objective response rates of up to 50% in patients with certain cancers. However, the significant number of patients and cancer types that altogether fail or acquire resistance to these therapies highlights the need for novel immunotherapies that target alternate pathways and effector cells. Thus, there is renewed interest in harnessing the tumor-killing abilities of Natural Killer (NK) cells, though it has proven difficult to efficiently and specifically target these cells cancer patients. The commercial success of T cell checkpoint inhibitors has seen a swam of new biotech companies emerge with innovative or revised strategies that aim to harness the innate non-antigen dependent tumor lysis potential of NK cells. This review will focus on IL-15 biology in NK cells and proposes the development novel therapies aimed at this pathway in humans.
Collapse
Affiliation(s)
- Jai Rautela
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Victoria 3010, Australia
| | - Nicholas D Huntington
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Victoria 3010, Australia.
| |
Collapse
|
49
|
Yuan ZX, Mo J, Zhao G, Shu G, Fu HL, Zhao W. Targeting Strategies for Renal Cell Carcinoma: From Renal Cancer Cells to Renal Cancer Stem Cells. Front Pharmacol 2016; 7:423. [PMID: 27891093 PMCID: PMC5103413 DOI: 10.3389/fphar.2016.00423] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 10/25/2016] [Indexed: 12/14/2022] Open
Abstract
Renal cell carcinoma (RCC) is a common form of urologic tumor that originates from the highly heterogeneous epithelium of renal tubules. Over the last decade, targeting therapies to renal cancer cells have transformed clinical care for RCC. Recently, it was proposed that renal cancer stem cells (CSCs) isolated from renal carcinomas were responsible for driving tumor growth and resistance to conventional chemotherapy and radiotherapy, according to the theory of CSCs; this has provided the rationale for therapies targeting this aggressive cell population. Precise identification of renal CSC populations and the complete cell hierarchy will accurately inform characterization of disease subtypes. This will ultimately contribute to more personalized and targeted therapies. Here, we summarize potential targeting strategies for renal cancer cells and renal CSCs, including tyrosine kinase inhibitors, mammalian target of rapamycin inhibitors (mTOR), interleukins, CSC marker inhibitors, bone morphogenetic protein-2, antibody drug conjugates, and nanomedicine. In conclusion, targeting therapies for RCC represent new directions for exploration and clinical investigation and they plant a seed of hope for advanced clinical care.
Collapse
Affiliation(s)
- Zhi-Xiang Yuan
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University Chengdu, China
| | - Jingxin Mo
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen UniversityGuangzhou, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen UniversityGuangzhou, China
| | - Guixian Zhao
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University Chengdu, China
| | - Gang Shu
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University Chengdu, China
| | - Hua-Lin Fu
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University Chengdu, China
| | - Wei Zhao
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen UniversityGuangzhou, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen UniversityGuangzhou, China
| |
Collapse
|
50
|
Affiliation(s)
- Paolo Lissoni
- Department of Oncological Service, Institute of Biological Medicine, Milan, Italy
| |
Collapse
|