51
|
Yu EM, Cho E, Singh R, Kim SH, Han C, Han S, Lee DG, Kim YH, Kwon BS, Choi BK. IL7-Fc Enhances the Efficacy of Adoptive T Cell Therapy under Lymphopenic Conditions in a Murine Melanoma Model. Cells 2021; 10:2018. [PMID: 34440787 PMCID: PMC8392867 DOI: 10.3390/cells10082018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 07/29/2021] [Accepted: 08/05/2021] [Indexed: 12/04/2022] Open
Abstract
Adoptive cell therapy (ACT) using tumor-reactive T cells is a promising form of immunotherapy to specifically target cancer. However, the survival and functional maintenance of adoptively transferred T cells remains a challenge, ultimately limiting their efficacy. Here, we evaluated the use of recombinant IL7-Fc in ACT. In a lymphopenic murine melanoma model, IL7-Fc treatment led to the enhanced inhibition of tumor growth with an increased number of adoptively transferred CD8+ T cells in tumor tissue and tumor-draining lymph nodes. Additionally, IL7-Fc further enhanced anti-tumor responses that were induced by recombinant human IL2 in the same mouse model. In contrast, in an immunocompetent murine melanoma model, IL7-Fc dampened the anti-tumor immunity. Further, IL7-Fc decreased the proliferation of adoptively transferred and immune-activated tumor-reactive CD8+ T cells in immunocompetent mice by inducing the massive expansion of endogenous T cells, thereby limiting the space for adoptively transferred T cells. Our data suggest that IL7-Fc is principally beneficial for enhancing the efficacy of tumor-reactive T-cells in lymphopenic conditions for the ACT.
Collapse
Affiliation(s)
- Eun M. Yu
- Biomedicine Production Branch, Program for Immunotherapy Research, National Cancer Center, Goyang 10408, Korea; (E.M.Y.); (D.G.L.); (Y.H.K.)
| | - Eunjung Cho
- Division of Tumor Immunology, National Cancer Center, Goyang 10408, Korea; (E.C.); (R.S.); (S.-H.K.); (C.H.); (S.H.)
| | - Rohit Singh
- Division of Tumor Immunology, National Cancer Center, Goyang 10408, Korea; (E.C.); (R.S.); (S.-H.K.); (C.H.); (S.H.)
| | - Seon-Hee Kim
- Division of Tumor Immunology, National Cancer Center, Goyang 10408, Korea; (E.C.); (R.S.); (S.-H.K.); (C.H.); (S.H.)
| | - Chungyong Han
- Division of Tumor Immunology, National Cancer Center, Goyang 10408, Korea; (E.C.); (R.S.); (S.-H.K.); (C.H.); (S.H.)
| | - Seongeun Han
- Division of Tumor Immunology, National Cancer Center, Goyang 10408, Korea; (E.C.); (R.S.); (S.-H.K.); (C.H.); (S.H.)
| | - Don G. Lee
- Biomedicine Production Branch, Program for Immunotherapy Research, National Cancer Center, Goyang 10408, Korea; (E.M.Y.); (D.G.L.); (Y.H.K.)
| | - Young H. Kim
- Biomedicine Production Branch, Program for Immunotherapy Research, National Cancer Center, Goyang 10408, Korea; (E.M.Y.); (D.G.L.); (Y.H.K.)
- Eutilex, Co., Ltd., Geumcheon-gu, Seoul 08594, Korea;
| | - Byoung S. Kwon
- Eutilex, Co., Ltd., Geumcheon-gu, Seoul 08594, Korea;
- Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
| | - Beom K. Choi
- Biomedicine Production Branch, Program for Immunotherapy Research, National Cancer Center, Goyang 10408, Korea; (E.M.Y.); (D.G.L.); (Y.H.K.)
| |
Collapse
|
52
|
Bonte S, de Munter S, Billiet L, Goetgeluk G, Ingels J, Jansen H, Pille M, de Cock L, Weening K, Taghon T, Leclercq G, Vandekerckhove B, Kerre T. In vitro OP9-DL1 co-culture and subsequent maturation in the presence of IL-21 generates tumor antigen-specific T cells with a favorable less-differentiated phenotype and enhanced functionality. Oncoimmunology 2021; 10:1954800. [PMID: 34367734 PMCID: PMC8312599 DOI: 10.1080/2162402x.2021.1954800] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
T cell receptor (TCR)-redirected T cells target intracellular antigens such as Wilms' tumor 1 (WT1), a tumor-associated antigen overexpressed in several malignancies, including acute myeloid leukemia (AML). For both chimeric antigen receptor (CAR)- and TCR-redirected T cells, several clinical studies indicate that T cell subsets with a less-differentiated phenotype (e.g. stem cell memory T cells, TSCM) survive longer and mediate superior anti-tumor effects in vivo as opposed to more terminally differentiated T cells. Cytokines added during in vitro and ex vivo culture of T cells play an important role in driving the phenotype of T cells for adoptive transfer. Using the OP9-DL1 co-culture system, we have shown previously that we are able to generate in vitro, starting from clinically relevant stem cell sources, T cells with a single tumor antigen (TA)-specific TCR. This method circumvents possible TCR chain mispairing and unwanted toxicities that might occur when introducing a TA-specific TCR in peripheral blood lymphocytes. We now show that we are able to optimize our in vitro culture protocol, by adding IL-21 during maturation, resulting in generation of TA-specific T cells with a less-differentiated phenotype and enhanced in vitro anti-tumor effects. We believe the favorable TSCM-like phenotype of these in vitro generated T cells preludes superior in vivo persistence and anti-tumor efficacy. Therefore, these TA-specific T cells could be of use as a valuable new form of patient-tailored T cell immunotherapy for malignancies for which finding a suitable CAR-T target antigen is challenging, such as AML.
Collapse
Affiliation(s)
- Sarah Bonte
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Stijn de Munter
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.,Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Lore Billiet
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Glenn Goetgeluk
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.,Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Joline Ingels
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Hanne Jansen
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Melissa Pille
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Laurenz de Cock
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Karin Weening
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Tom Taghon
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.,Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Georges Leclercq
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.,Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Bart Vandekerckhove
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.,Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Tessa Kerre
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium.,Department of Diagnostic Sciences, Ghent University, Ghent, Belgium.,Department of Hematology, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
53
|
Park JA, Santich BH, Xu H, Lum LG, Cheung NKV. Potent ex vivo armed T cells using recombinant bispecific antibodies for adoptive immunotherapy with reduced cytokine release. J Immunother Cancer 2021; 9:e002222. [PMID: 33986124 PMCID: PMC8126293 DOI: 10.1136/jitc-2020-002222] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND T cell-based immunotherapies using chimeric antigen receptors (CAR) or bispecific antibodies (BsAb) have produced impressive responses in hematological malignancies. However, major hurdles remained, including cytokine release syndrome, neurotoxicity, on-target off-tumor effects, reliance on autologous T cells, and failure in most solid tumors. BsAb armed T cells offer a safe alternative. METHODS We generated ex vivo armed T cells (EATs) using IgG-[L]-scFv-platformed BsAb, where the anti-CD3 (huOKT3) scFv was attached to the light chain of a tumor-binding IgG. BsAb density on EAT, in vitro cytotoxicity, cytokine release, in vivo trafficking into tumors, and their antitumor activities were evaluated in multiple cancer cell lines and patient-derived xenograft mouse models. The efficacy of EATs after cryopreservation was studied, and gamma delta (γδ) T cells were investigated as unrelated alternative effector T cells. RESULTS The antitumor potency of BsAb armed T cells was substantially improved using the IgG-[L]-scFv BsAb platform. When compared with separate BsAb and T cell injection, EATs released less TNF-α, and infiltrated tumors faster, while achieving robust antitumor responses. The in vivo potency of EAT therapy depended on BsAb dose for arming, EAT cell number per injection, total number of EAT doses, and treatment schedule intensity. The antitumor efficacy of EATs was preserved following cryopreservation, and EATs using γδ T cells were safe and as effective as αβ T cell-EATs. CONCLUSIONS EATs exerted potent antitumor activities against a broad spectrum of human cancer targets with remarkable safety. The antitumor potency of EATs depended on BsAb dose, cell number and total dose, and schedule. EATs were equally effective after cryopreservation, and the feasibility of third-party γδ-EATs offered an alternative for autologous T cell sources.
Collapse
MESH Headings
- Animals
- Antibodies, Bispecific/genetics
- Antibodies, Bispecific/immunology
- Antibodies, Bispecific/metabolism
- Cell Line, Tumor
- Cell Movement
- Coculture Techniques
- Cytokines/metabolism
- Cytotoxicity, Immunologic
- Humans
- Immunotherapy, Adoptive
- Intraepithelial Lymphocytes/immunology
- Intraepithelial Lymphocytes/metabolism
- Intraepithelial Lymphocytes/transplantation
- Lymphocyte Activation
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/transplantation
- Male
- Mice, Inbred BALB C
- Mice, Knockout
- Neoplasms/genetics
- Neoplasms/immunology
- Neoplasms/metabolism
- Neoplasms/therapy
- Phenotype
- Tumor Microenvironment
- Xenograft Model Antitumor Assays
- Mice
Collapse
Affiliation(s)
- Jeong A Park
- Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Brian H Santich
- Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Hong Xu
- Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Lawrence G Lum
- Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Nai-Kong V Cheung
- Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
54
|
Patidar M, Yadav N, Dalai SK. Development of Stable Chimeric IL-15 for Trans-Presentation by the Antigen Presenting Cells. Front Immunol 2021; 12:646159. [PMID: 33953717 PMCID: PMC8092395 DOI: 10.3389/fimmu.2021.646159] [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: 12/25/2020] [Accepted: 03/15/2021] [Indexed: 11/23/2022] Open
Abstract
IL-15 is one of the important biologics considered for vaccine adjuvant and treatment of cancer. However, a short half-life and poor bioavailability limit its therapeutic potential. Herein, we have structured IL-15 into a chimeric protein to improve its half-life enabling greater bioavailability for longer periods. We have covalently linked IL-15 with IgG2 base to make the IL-15 a stable chimeric protein, which also increased its serum half-life by 40 fold. The dimeric structure of this kind of IgG based biologics has greater stability, resistance to proteolytic cleavage, and less frequent dosing schedule with minimum dosage for achieving the desired response compared to that of their monomeric forms. The structured chimeric IL-15 naturally forms a dimer, and retains its affinity for binding to its receptor, IL-15Rβ. Moreover, with the focused action of the structured chimeric IL-15, antigen-presenting cells (APC) would transpresent chimeric IL-15 along with antigen to the T cell, that will help the generation of quantitatively and qualitatively better antigen-specific memory T cells. In vitro and in vivo studies demonstrate the biological activity of chimeric IL-15 with respect to its ability to induce IL-15 signaling and modulating CD8+ T cell response in favor of memory generation. Thus, a longer half-life, dimeric nature, and anticipated focused transpresentation by APCs to the T cells will make chimeric IL-15 a super-agonist for memory CD8+ T cell responses.
Collapse
Affiliation(s)
- Manoj Patidar
- Institute of Science, Nirma University, Ahmedabad, India.,Department of Zoology, Govt. College Manawar, Dhar, India
| | - Naveen Yadav
- Institute of Science, Nirma University, Ahmedabad, India.,Translation Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, India
| | - Sarat K Dalai
- Institute of Science, Nirma University, Ahmedabad, India
| |
Collapse
|
55
|
Anti-PD-1/PD-L1 Based Combination Immunotherapy to Boost Antigen-Specific CD8 + T Cell Response in Hepatocellular Carcinoma. Cancers (Basel) 2021; 13:cancers13081922. [PMID: 33923463 PMCID: PMC8073815 DOI: 10.3390/cancers13081922] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/22/2021] [Accepted: 04/14/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary The cytotoxic T cell response against hepatocellular carcinoma antigens is exhausted and fails in its task of deleting tumoral cells. These cells are featured by the expression of negative immune checkpoints that can be modulated to restore T cell function. The blockade of the PD-1/PD-L1 pathway has shown promising results in rescuing hepatocellular carcinoma-specific CD8 T cells but only a reduced group of cases is sensitive to this treatment and the effect is usually temporary. Therefore, new anti-PD-1 based combinatory strategies are underway to increase the response by adding the effect of blocking neo-angiogenesis and other negative immune checkpoints, boosting positive immune checkpoints, blocking suppressive cytokines, or inducing the expression of tumoral neoantigens. The restoration of T cell responses with these anti-PD-1 based combinatory therapies will change the outcome of advanced hepatocellular carcinoma. Abstract Thirty to fifty percent of hepatocellular carcinomas (HCC) display an immune class genetic signature. In this type of tumor, HCC-specific CD8 T cells carry out a key role in HCC control. Those potential reactive HCC-specific CD8 T cells recognize either HCC immunogenic neoantigens or aberrantly expressed host’s antigens, but they become progressively exhausted or deleted. These cells express the negative immunoregulatory checkpoint programmed cell death protein 1 (PD-1) which impairs T cell receptor signaling by blocking the CD28 positive co-stimulatory signal. The pool of CD8 cells sensitive to anti-PD-1/PD-L1 treatment is the PD-1dim memory-like precursor pool that gives rise to the effector subset involved in HCC control. Due to the epigenetic imprints that are transmitted to the next generation, the effect of PD-1 blockade is transient, and repeated treatments lead to tumor resistance. During long-lasting disease, besides the TCR signaling impairment, T cells develop other failures that should be also set-up to increase T cell reactivity. Therefore, several PD-1 blockade-based combinatory therapies are currently under investigation such as adding antiangiogenics, anti-TGFβ1, blockade of other negative immune checkpoints, or increasing HCC antigen presentation. The effect of these combinations on CD8+ T cells is discussed in this review.
Collapse
|
56
|
Cui X, Han L, Liu Y, Li Y, Sun W, Song B, Zhou W, Zhang Y, Wang H. siGCD: a web server to explore survival interaction of genes, cells and drugs in human cancers. Brief Bioinform 2021; 22:6210070. [PMID: 33822887 DOI: 10.1093/bib/bbab058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 01/19/2021] [Accepted: 02/12/2021] [Indexed: 11/13/2022] Open
Abstract
It is pivotal and remains challenge for cancer precision treatment to identify the survival outcome interactions between genes, cells and drugs. Here, we present siGCD, a web-based tool for analysis and visualization of the survival interaction of Genes, Cells and Drugs in human cancers. siGCD utilizes the cancer heterogeneity to simulate the manipulated gene expression, cell infiltration and drug treatment, which overcomes the data and experimental limitations. To illustrate the performance of siGCD, we identified the survival interaction partners of EGFR (gene level), T cells (cell level) and sorafenib (drug level), and our prediction was consistent with previous reports. Moreover, we validate the synergistic effect of regorafenib and glyburide, and found that glyburide could significantly improve the regorafenib response. These results demonstrate that siGCD could benefit cancer precision medicine in a wide range of advantageous application scenarios including gene regulatory network construction, immune cell regulatory gene identification, drug (especially multiple target drugs) response biomarker screening and combination therapeutic design.
Collapse
Affiliation(s)
- XiuLiang Cui
- National Center for Liver Cancer, the Second Military Medical University, Shanghai 200438, China
| | - Lu Han
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing China
| | - Yang Liu
- Institute for Hepatology, The Second Affiliated Hospital, South University of Science and Technology, 29 Bulan Road, Longgang District, Shenzhen 518112, China
| | - Ying Li
- Department of Pharmacy, Eastern Hepatobiliary Surgery Hospital, the Second Military Medical University, Shanghai 200438, China
| | - Wen Sun
- National Center for Liver Cancer, the Second Military Medical University, Shanghai 200438, China
| | - Bin Song
- Department of Pancreatic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200438, China
| | - Wenxia Zhou
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing China
| | - Yongxiang Zhang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing China
| | - Hongyang Wang
- National Center for Liver Cancer, the Second Military Medical University, Shanghai 200438, China.,International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital/Institute, the Second Military Medical University, 200438, Shanghai, China
| |
Collapse
|
57
|
Swadling L, Pallett LJ, Diniz MO, Baker JM, Amin OE, Stegmann KA, Burton AR, Schmidt NM, Jeffery-Smith A, Zakeri N, Suveizdyte K, Froghi F, Fusai G, Rosenberg WM, Davidson BR, Schurich A, Simon AK, Maini MK. Human Liver Memory CD8 + T Cells Use Autophagy for Tissue Residence. Cell Rep 2021; 30:687-698.e6. [PMID: 31968246 PMCID: PMC6988113 DOI: 10.1016/j.celrep.2019.12.050] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 11/04/2019] [Accepted: 12/13/2019] [Indexed: 12/11/2022] Open
Abstract
Tissue-resident memory T cells have critical roles in long-term pathogen and tumor immune surveillance in the liver. We investigate the role of autophagy in equipping human memory T cells to acquire tissue residence and maintain functionality in the immunosuppressive liver environment. By performing ex vivo staining of freshly isolated cells from human liver tissue, we find that an increased rate of basal autophagy is a hallmark of intrahepatic lymphocytes, particularly liver-resident CD8+ T cells. CD8+ T cells with increased autophagy are those best able to proliferate and mediate cytotoxicity and cytokine production. Conversely, blocking autophagy induction results in the accumulation of depolarized mitochondria, a feature of exhausted T cells. Primary hepatic stellate cells or the prototypic hepatic cytokine interleukin (IL)-15 induce autophagy in parallel with tissue-homing/retention markers. Inhibition of T cell autophagy abrogates tissue-residence programming. Thus, upregulation of autophagy adapts CD8+ T cells to combat mitochondrial depolarization, optimize functionality, and acquire tissue residence. An increased rate of basal autophagy is a hallmark of liver-resident CD8+ T cells Enhanced T cell autophagy can be imprinted by IL-15 or hepatic stellate cells Autophagy induction is required for tissue-residence programming in vitro Enhanced autophagy maintains TRM mitochondrial fitness in the liver
Collapse
Affiliation(s)
- Leo Swadling
- Division of Infection and Immunity, University College London, London, UK.
| | - Laura J Pallett
- Division of Infection and Immunity, University College London, London, UK
| | - Mariana O Diniz
- Division of Infection and Immunity, University College London, London, UK
| | - Josephine M Baker
- Division of Infection and Immunity, University College London, London, UK
| | - Oliver E Amin
- Division of Infection and Immunity, University College London, London, UK
| | - Kerstin A Stegmann
- Division of Infection and Immunity, University College London, London, UK
| | - Alice R Burton
- Division of Infection and Immunity, University College London, London, UK
| | - Nathalie M Schmidt
- Division of Infection and Immunity, University College London, London, UK
| | - Anna Jeffery-Smith
- Division of Infection and Immunity, University College London, London, UK; Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and Dentistry, QMUL, London, UK
| | - Nekisa Zakeri
- Division of Infection and Immunity, University College London, London, UK
| | | | - Farid Froghi
- Institute for Liver and Digestive Health, University College London, London, UK
| | - Giuseppe Fusai
- Institute for Liver and Digestive Health, University College London, London, UK
| | - William M Rosenberg
- Institute for Liver and Digestive Health, University College London, London, UK
| | - Brian R Davidson
- Institute for Liver and Digestive Health, University College London, London, UK
| | - Anna Schurich
- Division of Infection and Immunity, University College London, London, UK; Department of Infectious Diseases, Kings College London, London, UK
| | - A Katharina Simon
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
| | - Mala K Maini
- Division of Infection and Immunity, University College London, London, UK.
| |
Collapse
|
58
|
Marton C, Mercier-Letondal P, Galaine J, Godet Y. An unmet need: Harmonization of IL-7 and IL-15 combination for the ex vivo generation of minimally differentiated T cells. Cell Immunol 2021; 363:104314. [PMID: 33677140 DOI: 10.1016/j.cellimm.2021.104314] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 02/06/2021] [Accepted: 02/07/2021] [Indexed: 11/29/2022]
Abstract
T cell-based adoptive cell transfer therapy is now clinically used to fight cancer with CD19-targeting chimeric antigen receptor T cells. The use of other T cell-based immunotherapies relying on antigen-specific T cells, genetically modified or not, is expanding in various neoplastic diseases. T cell manufacturing has evolved through sophisticated processes to produce T cells with improved therapeutic potential. Clinical-grade manufacturing processes associated with these therapies must meet pharmaceutical requirements and therefore be standardized. Here, we focus on the use of cytokines to expand minimally differentiated T cells, as well as their standardization and harmonization in research and clinical settings.
Collapse
Affiliation(s)
- Chrystel Marton
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besançon, France.
| | - Patricia Mercier-Letondal
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besançon, France
| | - Jeanne Galaine
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besançon, France
| | - Yann Godet
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besançon, France.
| |
Collapse
|
59
|
Kadiyala P, Carney SV, Gauss JC, Garcia-Fabiani MB, Haase S, Alghamri MS, Núñez FJ, Liu Y, Yu M, Taher A, Nunez FM, Li D, Edwards MB, Kleer CG, Appelman H, Sun Y, Zhao L, Moon JJ, Schwendeman A, Lowenstein PR, Castro MG. Inhibition of 2-hydroxyglutarate elicits metabolic reprogramming and mutant IDH1 glioma immunity in mice. J Clin Invest 2021; 131:139542. [PMID: 33332283 DOI: 10.1172/jci139542] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 12/09/2020] [Indexed: 02/06/2023] Open
Abstract
Mutant isocitrate dehydrogenase 1 (IDH1-R132H; mIDH1) is a hallmark of adult gliomas. Lower grade mIDH1 gliomas are classified into 2 molecular subgroups: 1p/19q codeletion/TERT-promoter mutations or inactivating mutations in α-thalassemia/mental retardation syndrome X-linked (ATRX) and TP53. This work focuses on glioma subtypes harboring mIDH1, TP53, and ATRX inactivation. IDH1-R132H is a gain-of-function mutation that converts α-ketoglutarate into 2-hydroxyglutarate (D-2HG). The role of D-2HG within the tumor microenvironment of mIDH1/mATRX/mTP53 gliomas remains unexplored. Inhibition of D-2HG, when used as monotherapy or in combination with radiation and temozolomide (IR/TMZ), led to increased median survival (MS) of mIDH1 glioma-bearing mice. Also, D-2HG inhibition elicited anti-mIDH1 glioma immunological memory. In response to D-2HG inhibition, PD-L1 expression levels on mIDH1-glioma cells increased to similar levels as observed in WT-IDH gliomas. Thus, we combined D-2HG inhibition/IR/TMZ with anti-PDL1 immune checkpoint blockade and observed complete tumor regression in 60% of mIDH1 glioma-bearing mice. This combination strategy reduced T cell exhaustion and favored the generation of memory CD8+ T cells. Our findings demonstrate that metabolic reprogramming elicits anti-mIDH1 glioma immunity, leading to increased MS and immunological memory. Our preclinical data support the testing of IDH-R132H inhibitors in combination with IR/TMZ and anti-PDL1 as targeted therapy for mIDH1/mATRX/mTP53 glioma patients.
Collapse
Affiliation(s)
- Padma Kadiyala
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Stephen V Carney
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Jessica C Gauss
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Maria B Garcia-Fabiani
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Santiago Haase
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Mahmoud S Alghamri
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Felipe J Núñez
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Yayuan Liu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan, USA
| | - Minzhi Yu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan, USA
| | - Ayman Taher
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Fernando M Nunez
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Dan Li
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan, USA
| | - Marta B Edwards
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Celina G Kleer
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Henry Appelman
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Yilun Sun
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Biostatistics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Lili Zhao
- Department of Biostatistics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan, USA.,Biointerfaces Institute, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan, USA.,Biointerfaces Institute, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Pedro R Lowenstein
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Biointerfaces Institute, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Maria G Castro
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Biointerfaces Institute, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
60
|
Bernard PL, Laletin V, Pastor S, Nunès JA, Guittard G. [Unleashing NK cell signaling to improve cancer immunotherapy]. Med Sci (Paris) 2020; 36 Hors série n° 1:50-55. [PMID: 33052095 DOI: 10.1051/medsci/2020195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Pierre-Louis Bernard
- Centre de Recherche en Cancérologie de Marseille, CRCM, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille Université, Marseille, France
| | - Vladimir Laletin
- Centre de Recherche en Cancérologie de Marseille, CRCM, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille Université, Marseille, France
| | - Sonia Pastor
- Centre de Recherche en Cancérologie de Marseille, CRCM, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille Université, Marseille, France
| | - Jacques A Nunès
- Centre de Recherche en Cancérologie de Marseille, CRCM, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille Université, Marseille, France
| | - Geoffrey Guittard
- Centre de Recherche en Cancérologie de Marseille, CRCM, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille Université, Marseille, France
| |
Collapse
|
61
|
Chen P, Chen H, Moussa M, Cheng J, Li T, Qin J, Lifson JD, Sneller MC, Krymskaya L, Godin S, Lane HC, Catalfamo M. Recombinant Human Interleukin-15 and Anti-PD-L1 Combination Therapy Expands a CXCR3+PD1-/low CD8 T-Cell Subset in Simian Immunodeficiency Virus-Infected Rhesus Macaques. J Infect Dis 2020; 221:523-533. [PMID: 31562760 DOI: 10.1093/infdis/jiz485] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 09/23/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The PD1/PD-L1 pathway contributes to the pathogenesis of human immunodeficiency virus (HIV)/simian immunodeficiency virus (SIV) infection, and blockade of this pathway may have potential to restore immune function and promote viral control or elimination. In this study, we combined a checkpoint inhibitor anti-PD-L1 (Avelumab) and recombinant human interleukin-15 (rhIL-15) in SIV-infected rhesus macaques (RM). METHODS The rhIL-15 was administered as continuous infusion in 2 cycles of 10 days in the context of weekly administration of anti-PD-L1 (Avelumab) in SIV-infected RM receiving combination antiretroviral therapy (cART). Safety, immunological parameters, and viral loads were monitored during the study. RESULTS Administration of rhIL-15/anti-PD-L1 was safe and well tolerated. Treatment resulted in transient increases in proliferating (Ki67+) natural killer and CD8 T cells. In addition, treatment expanded a CXCR3+PD1-/low CD8 T-cell subset with the ability to secrete cytokines. Despite these effects, no changes in plasma viremia were observed after cART interruption. CONCLUSIONS Expansion of the CXCR3+PD1-/low CD8 T-cell subset with functional capacity and potential to traffic to sites of viral reservoirs in SIV-infected rhesus macaques had no demonstrable effect on plasma viremia after cART interruption.
Collapse
Affiliation(s)
- Ping Chen
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington District of Columbia, USA
| | - Hui Chen
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington District of Columbia, USA.,CMRS/Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Maha Moussa
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington District of Columbia, USA
| | - Jie Cheng
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington District of Columbia, USA
| | - Tong Li
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington District of Columbia, USA
| | - Jing Qin
- Biostatistics Research Branch, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland, USA
| | - Michael C Sneller
- CMRS/Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Ludmila Krymskaya
- Clinical Support Laboratory, Leidos Biomedical Research, Inc., Frederick, Maryland, USA
| | - Steven Godin
- Smithers Avanza Toxicology Services, Gaithersburg, Maryland, USA
| | - H Clifford Lane
- CMRS/Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Marta Catalfamo
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington District of Columbia, USA
| |
Collapse
|
62
|
Mondino A, Manzo T. To Remember or to Forget: The Role of Good and Bad Memories in Adoptive T Cell Therapy for Tumors. Front Immunol 2020; 11:1915. [PMID: 32973794 PMCID: PMC7481451 DOI: 10.3389/fimmu.2020.01915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 07/16/2020] [Indexed: 12/17/2022] Open
Abstract
The generation of immunological memory is a hallmark of adaptive immunity by which the immune system "remembers" a previous encounter with an antigen expressed by pathogens, tumors, or normal tissues; and, upon secondary encounters, mounts faster and more effective recall responses. The establishment of T cell memory is influenced by both cell-intrinsic and cell-extrinsic factors, including genetic, epigenetic and environmental triggers. Our current knowledge of the mechanisms involved in memory T cell differentiation has instructed new opportunities to engineer T cells with enhanced anti-tumor activity. The development of adoptive T cell therapy has emerged as a powerful approach to cure a subset of patients with advanced cancers. Efficacy of this approach often requires long-term persistence of transferred T cell products, which can vary according to their origin and manufacturing conditions. Host preconditioning and post-transfer supporting strategies have shown to promote their engraftment and survival by limiting the competition with a hostile tumor microenvironment and between pre-existing immune cell subsets. Although in the general view pre-existing memory can confer a selective advantage to adoptive T cell therapy, here we propose that also "bad memories"-in the form of antigen-experienced T cell subsets-co-evolve with consequences on newly transferred lymphocytes. In this review, we will first provide an overview of selected features of memory T cell subsets and, then, discuss their putative implications for adoptive T cell therapy.
Collapse
Affiliation(s)
- Anna Mondino
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Teresa Manzo
- Department of Experimental Oncology, IRCCS European Institute of Oncology, Milan, Italy
| |
Collapse
|
63
|
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: 78] [Impact Index Per Article: 15.6] [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
|
64
|
Chen H, Moussa M, Catalfamo M. The Role of Immunomodulatory Receptors in the Pathogenesis of HIV Infection: A Therapeutic Opportunity for HIV Cure? Front Immunol 2020; 11:1223. [PMID: 32714317 PMCID: PMC7343933 DOI: 10.3389/fimmu.2020.01223] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 05/15/2020] [Indexed: 12/12/2022] Open
Abstract
Immune activation is the hallmark of HIV infection and plays a role in the pathogenesis of the disease. In the context of suppressed HIV RNA replication by combination antiretroviral therapy (cART), there remains immune activation which is associated to the HIV reservoirs. Persistent virus contributes to a sustained inflammatory environment promoting accumulation of "activated/exhausted" T cells with diminished effector function. These T cells show increased expression of immunomodulatory receptors including Programmed cell death protein (PD1), Cytotoxic T Lymphocyte Associated Protein 4 (CTLA4), Lymphocyte activation gene 3 (LAG3), T cell immunoglobulin and ITIM domain (TIGIT), T cell immunoglobulin and mucin domain containing 3 (TIM3) among others. More importantly, recent reports had demonstrated that, HIV infected T cells express checkpoint receptors, contributing to their survival and promoting maintenance of the viral reservoir. Therapeutic strategies are focused on viral reservoir elimination and/or those to achieve sustained cART-free virologic remission. In this review, we will discuss the immunological basis and the latest advances of the use of checkpoint inhibitors to treat HIV infection.
Collapse
Affiliation(s)
- Hui Chen
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington, DC, United States
- CMRS/Laboratory of Immunoregulation, National Institutes of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - Maha Moussa
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington, DC, United States
| | - Marta Catalfamo
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington, DC, United States
| |
Collapse
|
65
|
Ma Y, Ou J, Lin T, Chen L, Wang J, Qiao D, Lai S, Duan C, Cheng Y, Chang R, Zhang C, Wang M. Phenotypic analysis of tumor-infiltrating lymphocytes from non-small cell lung cancer and their potential application for adoptive cell therapy. Immunopharmacol Immunotoxicol 2020; 42:319-329. [PMID: 32419542 DOI: 10.1080/08923973.2020.1765375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
AIM Adoptive cell therapy (ACT) of tumor-infiltrating lymphocytes (TILs) has demonstrated clinical benefits in metastatic melanoma treatment. However, the clinical application of TILs produced by a widely used standard protocol from non-small cell lung cancer (NSCLC) can be quite challenging because of the limited clinical benefits. A comprehensive phenotypic knowledge of TILs obtained from NSCLC is important for the development and improvement of personalized TIL therapy for NSCLC patients. METHODS In this study, we successfully expanded TILs from 141 NSCLC tissues which can be used in clinical ACT after expansion by a rapid expansion protocol (REP). RESULTS Our study indicates that the clinicopathological characteristics of patients have considerable impacts on the phenotype of in vitro TIL culture products. Different culture conditions are necessary for patients with different clinical features. Specific manipulations before REP expansion are required depending on the different phenotypes of TIL cultures (e.g. depletion of immune-suppressive γδT cells). With these optimizations, next-generation TIL therapy may become a treatment alternative for NSCLC patients in the future.
Collapse
Affiliation(s)
- Yipeng Ma
- Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen International Biological Valley-Life Science Industrial Park, Shenzhen, PR China
| | - Jiayu Ou
- Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen International Biological Valley-Life Science Industrial Park, Shenzhen, PR China
| | - Tong Lin
- Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen International Biological Valley-Life Science Industrial Park, Shenzhen, PR China
| | - Lei Chen
- Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen International Biological Valley-Life Science Industrial Park, Shenzhen, PR China
| | - Juntao Wang
- Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen International Biological Valley-Life Science Industrial Park, Shenzhen, PR China
| | - Dongjuan Qiao
- Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen International Biological Valley-Life Science Industrial Park, Shenzhen, PR China
| | - Shuoyan Lai
- Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen International Biological Valley-Life Science Industrial Park, Shenzhen, PR China
| | - Chaojun Duan
- Department of Thoracic Surgery, Xiangya Hospital of Central South University, Changsha, PR China
| | - Yuanda Cheng
- Department of Thoracic Surgery, Xiangya Hospital of Central South University, Changsha, PR China
| | - Ruimin Chang
- Department of Thoracic Surgery, Xiangya Hospital of Central South University, Changsha, PR China
| | - Chunfang Zhang
- Department of Thoracic Surgery, Xiangya Hospital of Central South University, Changsha, PR China
| | - Mingjun Wang
- Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen International Biological Valley-Life Science Industrial Park, Shenzhen, PR China
| |
Collapse
|
66
|
Identification of a Gene-Related Risk Signature in Melanoma Patients Using Bioinformatic Profiling. JOURNAL OF ONCOLOGY 2020; 2020:7526204. [PMID: 32411243 PMCID: PMC7206882 DOI: 10.1155/2020/7526204] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 01/21/2020] [Indexed: 01/15/2023]
Abstract
Introduction Gene signature has been used to predict prognosis in melanoma patients. Meanwhile, the efficacy of immunotherapy was correlated with particular genes expression or mutation. In this study, we systematically explored the gene expression pattern in the melanoma-immune microenvironment and its relationship with prognosis. Methods A cohort of 122 melanoma cases with whole-genome microarray expression data were enrolled from the Gene Expression Omnibus (GEO) database. The findings were validated using The Cancer Genome Atlas (TCGA) database. A principal component analysis (PCA), gene set enrichment analysis (GSEA), and gene oncology (GO) analysis were performed to explore the bioinformatic implications. Results Different gene expression patterns were identified according to the clinical stage. All eligible gene sets were analyzed, and the 8 genes (GPR87, KIT, SH3GL3, PVRL1, ATP1B1, CDAN1, FAU, and TNFSF14) with the greatest prognostic impact on melanoma. A gene-related risk signature was developed to distinguish patients with a high or low risk of an unfavorable outcome, and this signature was validated using the TCGA database. Furthermore, the prognostic significance of the signature between the classified subgroups was verified as an independent prognostic predictor of melanoma. Additionally, the low-risk melanoma patients presented an enhanced immune phenotype compared to that of the high-risk gene signature patients. Conclusions The gene pattern differences in melanoma were profiled, and a gene signature that could independently predict melanoma patients with a high risk of poor survival was established, highlighting the relationship between prognosis and the local immune response.
Collapse
|
67
|
Batra SA, Rathi P, Guo L, Courtney AN, Fleurence J, Balzeau J, Shaik RS, Nguyen TP, Wu MF, Bulsara S, Mamonkin M, Metelitsa LS, Heczey A. Glypican-3-Specific CAR T Cells Coexpressing IL15 and IL21 Have Superior Expansion and Antitumor Activity against Hepatocellular Carcinoma. Cancer Immunol Res 2020; 8:309-320. [PMID: 31953246 PMCID: PMC10765595 DOI: 10.1158/2326-6066.cir-19-0293] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 11/08/2019] [Accepted: 01/10/2020] [Indexed: 01/08/2023]
Abstract
Hepatocellular carcinoma (HCC) is the fourth most common cause of cancer-related death in the world, and curative systemic therapies are lacking. Chimeric antigen receptor (CAR)-expressing T cells induce robust antitumor responses in patients with hematologic malignancies but have limited efficacy in patients with solid tumors, including HCC. IL15 and IL21 promote T-cell expansion, survival, and function and can improve the antitumor properties of T cells. We explored whether transgenic expression of IL15 and/or IL21 enhanced glypican-3-CAR (GPC3-CAR) T cells' antitumor properties against HCC. We previously optimized the costimulation in GPC3-CARs and selected a second-generation GPC3-CAR incorporating a 4-1BB costimulatory endodomain (GBBz) for development. Here, we generated constructs encoding IL15, IL21, or both with GBBz (15.GBBz, 21.GBBz, and 21.15.GBBz, respectively) and examined the ability of transduced T cells to kill, produce effector cytokines, and expand in an antigen-dependent manner. We performed gene-expression and phenotypic analyses of GPC3-CAR T cells and CRISPR-Cas9 knockout of the TCF7 gene. Finally, we measured GPC3-CAR T-cell antitumor activity in murine xenograft models of GPC3+ tumors. The increased proliferation of 21.15.GBBz T cells was at least in part dependent on the upregulation and maintenance of TCF-1 (encoded by TCF7) and associated with a higher percentage of stem cell memory and central memory populations after manufacturing. T cells expressing 21.15.GBBz had superior in vitro and in vivo expansion and persistence, and the most robust antitumor activity in vivo These results provided preclinical evidence to support the clinical evaluation of 21.15.GPC3-CAR T cells in patients with HCC.
Collapse
Affiliation(s)
- Sai Arun Batra
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Purva Rathi
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Linjie Guo
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Amy N Courtney
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Julien Fleurence
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Julien Balzeau
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Rahamthulla S Shaik
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Thao P Nguyen
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Meng-Fen Wu
- Dan L Duncan Cancer Center Biostatistics Shared Resource, Baylor College of Medicine, Houston, Texas
| | - Shaun Bulsara
- Dan L Duncan Cancer Center Biostatistics Shared Resource, Baylor College of Medicine, Houston, Texas
| | - Maksim Mamonkin
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
| | - Leonid S Metelitsa
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
| | - Andras Heczey
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas.
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children's Hospital Liver Tumor Center, Houston, Texas
| |
Collapse
|
68
|
Pilipow K, Scamardella E, Lugli E. Generating stem-like memory T cells with antioxidants for adoptive cell transfer immunotherapy of cancer. Methods Enzymol 2019; 631:137-158. [PMID: 31948545 DOI: 10.1016/bs.mie.2019.08.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Among the multiple factors that are responsible for the success of adoptive cell transfer (ACT) immunotherapy for cancer, the differentiation status of the in vitro expanded T cell product at the time of transfer seems to play a major role. In particular, less differentiated memory CD8+ T cells endowed with self-renewing capacity and multipotency exert the most potent antitumor activity. To this aim, expansion protocols that generate sufficient numbers of tumor-specific CD8+ T cells with superior capacity to persist in vivo following ACT are needed. We describe a procedure for the differentiation of TCF-1+ stem-like CD8+ memory T cells from peripheral blood naïve precursors that takes advantage of the use of antioxidants, in particular N-acetylcysteine (NAC), in combination with T cell receptor stimulation and proinflammatory cytokines. We additionally describe how to conduct in vitro assays to test the stem-like features of the generated cells at the phenotypic, functional and metabolic level. Balancing the oxidative metabolism by the addition of antioxidants during in vitro manipulation of CD8+ T cells results in the generation of cell products with potent antitumor characteristics following ACT.
Collapse
Affiliation(s)
- Karolina Pilipow
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Eloise Scamardella
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Enrico Lugli
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy; Flow Cytometry Core, Humanitas Clinical and Research Center, Rozzano, Milan, Italy.
| |
Collapse
|
69
|
Pan Y, Wang Z, Zhang G, Guo J, Zhu X, Zhou J, Zhang Z, Sun Z, Yang J, Kastin AJ, Pan W, Wu X, Zhang J, Wang X, Wang C, He Y. Schizophrenia Patient Shows a Rare Interleukin 15 Receptor alpha Variant Disrupting Signal Transduction. Curr Mol Med 2019; 19:560-569. [PMID: 31244423 DOI: 10.2174/1566524019666190617172054] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 05/25/2019] [Accepted: 05/28/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Schizophrenia is a complex and debilitating mental disorder with strong heritability. Its pathogenesis involves immune dysregulation. Interleukin 15 and interleukin 15 receptor alpha(IL-15Rα) are classical immune molecules. They also help maintain normal brain function, leading to our hypothesis that IL-15Rα gene(IL- 15RA) variants contribute to the pathogenesis of schizophrenia. OBJECTIVE We determine whether the genetic variants of IL-15RA are associated with the development and progression of schizophrenia and whether IL-15RA single nucleotide polymorphism(SNP) plays a key role in downstream signaling transduction. METHODS AND RESULTS We sequenced IL-15RA exon from 132 Chinese schizophrenic patients and identified a rare variant(rs528238821) in a patient diagnosed with catatonic schizophrenia and ankylosing spondylitis(AS). We overexpressed this missense variant in cells driven by pBI-CMV vector. The cells showed attenuated STAT3 phosphorylation in response to interleukin15. CONCLUSION IL-15RA mutation is rare in schizophrenic patients but interfered with IL- 15Rα intracellular signal transduction. Given the similarity of symptoms of catatonic schizophrenia and the known phenotype of IL-15Rα knockout mice, gene variation might offer diagnostic value for sub-types of schizophrenia.
Collapse
Affiliation(s)
- Yanli Pan
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Zhimin Wang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Guangping Zhang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Junhua Guo
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Xuequan Zhu
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Jia Zhou
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Zhenrong Zhang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Zuoli Sun
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Jian Yang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Abba J Kastin
- Pennington Biomedical Research Center, Baton Rouge, LA70808, United States
| | - Weihong Pan
- BioPotentials Consulting, Sedona, AZ 86351, United States
| | - Xiaojun Wu
- Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Material Medical, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jianliang Zhang
- Department of Neurobiology, Capital Medical University, Beijing 100069, China.,Beijing Institute for Brain Disorders, Beijing 100069, China.,Beijing Center of Neural Regeneration and Repair, Beijing 100069, China
| | - Xiaomin Wang
- Beijing Institute for Brain Disorders, Beijing 100069, China.,Department of Physiology and Pathologic Physiology, Capital Medical University, Beijing 100069, China.,Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing 100069, China
| | - Chuanyue Wang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Yi He
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| |
Collapse
|
70
|
Kwak HW, Park HJ, Ko HL, Park H, Cha MH, Lee SM, Kang KW, Kim RH, Ryu SR, Kim HJ, Kim JO, Song M, Kim H, Jeong DG, Shin EC, Nam JH. Cricket paralysis virus internal ribosome entry site-derived RNA promotes conventional vaccine efficacy by enhancing a balanced Th1/Th2 response. Vaccine 2019; 37:5191-5202. [PMID: 31371226 PMCID: PMC7115557 DOI: 10.1016/j.vaccine.2019.07.070] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/21/2019] [Accepted: 07/18/2019] [Indexed: 12/20/2022]
Abstract
RNA adjuvant was developed from the CrPV intergenic region IRES. The RNA adjuvant functioned as an adjuvant with protein-based vaccines. The RNA adjuvant increased vaccine efficacy and induced balanced Th1/Th2 response. The RNA adjuvant enhanced APC chemotaxis.
An ideal adjuvant should increase vaccine efficacy through balanced Th1/Th2 responses and be safe to use. Recombinant protein-based vaccines are usually formulated with aluminum (alum)-based adjuvants to ensure an adequate immune response. However, use of alum triggers a Th2-biased immune induction, and hence is not optimal. Although the adjuvanticity of RNA has been reported, a systematic and overall investigation on its efficacy is lacking. We found that single strand RNA (termed RNA adjuvant) derived from cricket paralysis virus intergenic region internal ribosome entry site induced the expression of various adjuvant-function-related genes, such as type 1 and 2 interferon (IFN) and toll-like receptor (TLR), T cell activation, and leukocyte chemotaxis in human peripheral blood mononuclear cells; furthermore, its innate and IFN transcriptome profile patterns were similar to those of a live-attenuated yellow fever vaccine. This suggests that protein-based vaccines formulated using RNA adjuvant function as live-attenuated vaccines. Application of the RNA adjuvant in mouse enhanced the efficacy of Middle East respiratory syndrome spike protein, a protein-subunit vaccine and human papillomavirus L1 protein, a virus-like particle vaccine, by activating innate immune response through TLR7 and enhancing pAPC chemotaxis, leading to a balanced Th1/Th2 responses. Moreover, the combination of alum and the RNA adjuvant synergistically induced humoral and cellular immune responses and endowed long-term immunity. Therefore, RNA adjuvants have broad applicability and can be used with all conventional vaccines to improve vaccine efficacy qualitatively and quantitively.
Collapse
Affiliation(s)
- Hye Won Kwak
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Hyo-Jung Park
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Hae Li Ko
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Hyelim Park
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Min Ho Cha
- KM Application Center, Korea Institute of Oriental Medicine, Daegu, Republic of Korea
| | - Sang-Myeong Lee
- Division of Biotechnology, The Chonbuk National University, Iksan, Republic of Korea
| | - Kyung Won Kang
- Division of Biotechnology, The Chonbuk National University, Iksan, Republic of Korea
| | - Rhoon-Ho Kim
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Seung Rok Ryu
- Division of Biotechnology, The Chonbuk National University, Iksan, Republic of Korea
| | - Hye-Jung Kim
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Jae-Ouk Kim
- Clinical Research Lab, International Vaccine Institute, Seoul National University Research Park, Seoul, Republic of Korea
| | - Manki Song
- Clinical Research Lab, International Vaccine Institute, Seoul National University Research Park, Seoul, Republic of Korea
| | - Hun Kim
- Life Science Research Institute, SK Bioscience, Seongnam, Republic of Korea
| | - Dae Gwin Jeong
- Infectious Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Eui-Cheol Shin
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Jae-Hwan Nam
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Republic of Korea.
| |
Collapse
|
71
|
Brownlie RJ, Wright D, Zamoyska R, Salmond RJ. Deletion of PTPN22 improves effector and memory CD8+ T cell responses to tumors. JCI Insight 2019; 5:127847. [PMID: 31335326 PMCID: PMC6777814 DOI: 10.1172/jci.insight.127847] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Adoptive T cell therapy (ACT) has been established as an efficacious methodology for the treatment of cancer. Identifying targets to enhance the antigen recognition, functional capacity and longevity of T cells has the potential to broaden the applicability of these approaches in the clinic. We previously reported that targeting expression of phosphotyrosine phosphatase, non-receptor type (PTPN) 22 in effector CD8+ T cells enhances the efficacy of ACT for tumor clearance in mice. In the current work, we demonstrate that, upon ACT, PTPN22-deficient effector CD8+ T cells afford greater protection against tumors expressing very low affinity antigen, but do not survive long-term in vivo. Persistence of CD8+ T cells following tumor clearance is improved by ACT of memory phenotype cells that have a distinct metabolic phenotype as compared to effector T cells. Importantly, PTPN22-deficient T cells have comparable capacity to form long-lived memory cells in vivo but enhanced anti-tumor activity in vivo and effector responses ex vivo. These findings provide key insight into the regulation of effector and memory T cell responses in vivo, and indicate that PTPN22 is a rationale target to improve ACT for cancer.
Collapse
Affiliation(s)
- Rebecca J Brownlie
- Leeds Institute of Medical Research at St. James's, University of Leeds, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds, United Kingdom
| | - David Wright
- Institute of Immunology and Infection Research, University of Edinburgh, Ashworth Laboratories, Edinburgh, United Kingdom
| | - Rose Zamoyska
- Institute of Immunology and Infection Research, University of Edinburgh, Ashworth Laboratories, Edinburgh, United Kingdom
| | - Robert J Salmond
- Leeds Institute of Medical Research at St. James's, University of Leeds, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds, United Kingdom
| |
Collapse
|
72
|
Abstract
PURPOSE OF REVIEW Combination antiretroviral therapy (ART) has enabled tremendous progress in suppressing HIV replication in infected patients. However, ART alone cannot eradicate HIV and its latent, persisting reservoirs. Novel approaches are needed to eradicate the virus or achieve functional cure in the absence of ART. RECENT FINDINGS Adoptive T-cell therapies were initially tested in HIV-infected individuals with limited efficiency. Benefiting from new and improved methodologies, an increasing array of CAR T-cell therapies has been successfully developed in the cancer immunotherapy field, demonstrating promising new avenues that could be applied to HIV. Numerous studies have characterized various HIV-specific CAR constructs, types of cytolytic effector cells, and CAR-expressing cells' trafficking to the reservoir compartments, warranting further in-vivo efforts. Notably, the ability of CAR cells to persist and function in low-antigen environments in vivo, that is, in ART-suppressed patients, remains unclear. SUMMARY Despite promising results in preclinical studies, only a handful of clinical trials have been initiated worldwide. Several obstacles remain prior to successful application of HIV-specific CAR T-cell therapies in patients. In this review, we survey the current state of the field, and address paths towards realizing the goal of an efficacious HIV CAR T-cell product.
Collapse
|
73
|
Dwyer CJ, Knochelmann HM, Smith AS, Wyatt MM, Rangel Rivera GO, Arhontoulis DC, Bartee E, Li Z, Rubinstein MP, Paulos CM. Fueling Cancer Immunotherapy With Common Gamma Chain Cytokines. Front Immunol 2019; 10:263. [PMID: 30842774 PMCID: PMC6391336 DOI: 10.3389/fimmu.2019.00263] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 01/30/2019] [Indexed: 12/16/2022] Open
Abstract
Adoptive T cell transfer therapy (ACT) using tumor infiltrating lymphocytes or lymphocytes redirected with antigen receptors (CAR or TCR) has revolutionized the field of cancer immunotherapy. Although CAR T cell therapy mediates robust responses in patients with hematological malignancies, this approach has been less effective for treating patients with solid tumors. Additionally, toxicities post T cell infusion highlight the need for safer ACT protocols. Current protocols traditionally expand T lymphocytes isolated from patient tumors or from peripheral blood to large magnitudes in the presence of high dose IL-2 prior to infusion. Unfortunately, this expansion protocol differentiates T cells to a full effector or terminal phenotype in vitro, consequently reducing their long-term survival and antitumor effectiveness in vivo. Post-infusion, T cells face further obstacles limiting their persistence and function within the suppressive tumor microenvironment. Therapeutic manipulation of T cells with common γ chain cytokines, which are critical growth factors for T cells, may be the key to bypass such immunological hurdles. Herein, we discuss the primary functions of the common γ chain cytokines impacting T cell survival and memory and then elaborate on how these distinct cytokines have been used to augment T cell-based cancer immunotherapy.
Collapse
Affiliation(s)
- Connor J Dwyer
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Hannah M Knochelmann
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Aubrey S Smith
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Megan M Wyatt
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Guillermo O Rangel Rivera
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Dimitrios C Arhontoulis
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Eric Bartee
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Zihai Li
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Mark P Rubinstein
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Chrystal M Paulos
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
74
|
Guan L, Li X, Wei J, Liang Z, Yang J, Weng X, Wu X. Antigen-specific CD8+ memory stem T cells generated from human peripheral blood effectively eradicate allogeneic targets in mice. Stem Cell Res Ther 2018; 9:337. [PMID: 30526661 PMCID: PMC6286512 DOI: 10.1186/s13287-018-1080-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 11/12/2018] [Accepted: 11/19/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND As the implantation and long-term existence of tumor-specific T cells in host are the prerequisite for adoptive immunotherapy, memory stem T cells (TSCM) with self-renewal and differentiation capacity show the greatest potential to implant and long-term exhibit function in vivo, compared with other T cells of differentiation stages. Hence, tumor-specific TSCM have become potential candidate for adoptive T cell therapy of cancer. Here, we reported a protocol to generate allogeneic antigen-specific CD8+ TSCM cells from human PBLs. METHODS To prepare allogeneic antigen-specific CD8+ TSCM, we used an LCL named E007 of defined HLA allotyping as simulator, a co-culture of E007 and allogeneic PBLs was carried out in the presence of differentiation inhibitor TWS119 for 7 days. Sorting of proliferation cells ensured the E007-specificity of the prepared TSCM cells. The sorted lymphocytes underwent further expansion by cytokines IL-7 and IL-15 for further 7 days, making the E007-specific CD8 + TSCM expanded in number. The stem cell and T memory cell properties of the prepared CD8+ TSCM were observed in NOD-SCID mice. RESULTS Our protocol began with 1 × 107 PBLs and resulted in 2 × 107 E007-specific CD8+ TSCM cells in 2 weeks of preparation. The prepared TSCM cells exhibited a proliferative history and rapid differentiation into effector cells upon the E007 re-stimulation. Importantly, the prepared TSCM cells were able to exist long and reconstitute other T cell subsets in vivo, eradicating the E007 cells effectively after transferred into the LCL burden mice. CONCLUSIONS This protocol was able to prepare allogeneic antigen-specific CD8+ TSCM cells from human PBLs. The prepared TSCM showed the properties of stem cells and T memory cells. This study provided a reference method for generation of antigen-specific TSCM for T cell adoptive immunotherapy.
Collapse
Affiliation(s)
- Liping Guan
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Rd, Wuhan, 430030, China
| | - Xiaoyi Li
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Rd, Wuhan, 430030, China
| | - Jiali Wei
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Rd, Wuhan, 430030, China
| | - Zhihui Liang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Rd, Wuhan, 430030, China
| | - Jing Yang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Rd, Wuhan, 430030, China
| | - Xiufang Weng
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Rd, Wuhan, 430030, China.
| | - Xiongwen Wu
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Rd, Wuhan, 430030, China.
| |
Collapse
|
75
|
Duggal NA. Reversing the immune ageing clock: lifestyle modifications and pharmacological interventions. Biogerontology 2018; 19:481-496. [PMID: 30269199 PMCID: PMC6223743 DOI: 10.1007/s10522-018-9771-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 09/16/2018] [Indexed: 12/20/2022]
Abstract
It is widely accepted that ageing is accompanied by remodelling of the immune system, including reduced numbers of naïve T cells, increased senescent or exhausted T cells, compromise to monocyte, neutrophil and natural killer cell function and an increase in systemic inflammation. In combination these changes result in increased risk of infection, reduced immune memory, reduced immune tolerance and immune surveillance, with significant impacts upon health in old age. More recently it has become clear that the rate of decline in the immune system is malleable and can be influenced by environmental factors such as physical activity as well as pharmacological interventions. This review discusses briefly our current understanding of immunesenescence and then focuses on lifestyle interventions and therapeutic strategies that have been shown to restore immune functioning in aged individuals.
Collapse
Affiliation(s)
- Niharika A Duggal
- MRC-Arthritis Research UK Centre for Musculoskeletal Ageing Research, Institute of Inflammation and Ageing, Birmingham University, Birmingham, UK.
| |
Collapse
|
76
|
Liu L, Zong ZM, Liu Q, Jiang SS, Zhang Q, Cen LQ, Gao J, Gao XG, Huang JD, Liu Y, Yao H. A novel galactose-PEG-conjugated biodegradable copolymer is an efficient gene delivery vector for immunotherapy of hepatocellular carcinoma. Biomaterials 2018; 184:20-30. [PMID: 30195802 DOI: 10.1016/j.biomaterials.2018.08.064] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 08/30/2018] [Accepted: 08/31/2018] [Indexed: 12/20/2022]
Abstract
Successful immunogene therapy depends not only on the therapeutic gene but also on the gene delivery vector. In this study, we synthesized a novel copolymer consisting of low-molecular-weight polyethylenimine (PEI) cross-linked by myo-inositol (INO) and conjugated with a galactose-grafted PEG chain, named LA-PegPI. We characterized the chemical structure and molecular weight of the copolymer and particle properties of LA-PegPI/pDNA. Furthermore, we showed that LA-PegPI/pDNA polyplexes possessed excellent stability in physiological salt solution, low cytotoxicity, and high transfection efficiency in the asialoglycoprotein receptor (ASGPR)-positive liver cells in vitro. Importantly, we also showed that through intraperitoneal injection of LA-PegPI/pDNA nanoparticles, the reporter gene was forcefully expressed in the liver hepatocytes of mice. Finally, we documented that intraperitoneal injection of LA-PegPI/pIL15 nanoparticles effectively suppressed tumor growth and prolonged survival time of tumor-bearing mice via activation of CD8+ T cells and NK cells and upregulation of the cytokines IFN-γ, TNF, and IL12 in an orthotopic hepatocellular carcinoma mouse model. Interestingly, LA-PegPI/pluc nanoparticles could effectively stimulate the proliferation of NK cells and inhibit tumor growth in this model. In summary, LA-PegPI is a useful gene vector for immunogene therapy of hepatocellular carcinoma, and its potential for clinical application warrants further study.
Collapse
Affiliation(s)
- Ling Liu
- Key Laboratory of Coal Processing and Efficient Utilization, Ministry of Education, China University of Mining & Technology, Xuzhou, Jiangsu 221116, PR China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Zhi-Min Zong
- Key Laboratory of Coal Processing and Efficient Utilization, Ministry of Education, China University of Mining & Technology, Xuzhou, Jiangsu 221116, PR China.
| | - Qian Liu
- Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Shuang-Shuang Jiang
- Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Qian Zhang
- Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Lan-Qi Cen
- Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Jian Gao
- Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Xiao-Ge Gao
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Jian-Dong Huang
- Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, PR China; Department of Biochemistry and Shenzhen Institute of Research and Innovation, University of Hong Kong, 999077, Hong Kong, China
| | - Yi Liu
- Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Hong Yao
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, PR China.
| |
Collapse
|
77
|
Brummelman J, Mazza EMC, Alvisi G, Colombo FS, Grilli A, Mikulak J, Mavilio D, Alloisio M, Ferrari F, Lopci E, Novellis P, Veronesi G, Lugli E. High-dimensional single cell analysis identifies stem-like cytotoxic CD8 + T cells infiltrating human tumors. J Exp Med 2018; 215:2520-2535. [PMID: 30154266 PMCID: PMC6170179 DOI: 10.1084/jem.20180684] [Citation(s) in RCA: 261] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 07/06/2018] [Accepted: 08/16/2018] [Indexed: 01/17/2023] Open
Abstract
CD8+ T cells infiltrating tumors are largely dysfunctional, but whether a subset maintains superior functionality remains ill defined. By high-dimensional single cell analysis of millions of CD8+ T cells from 53 individuals with lung cancer, we defined those subsets that are enriched in tumors compared with cancer-free tissues and blood. Besides exhausted and activated cells, we identified CXCR5+ TIM-3- CD8+ T cells with a partial exhausted phenotype, while retaining gene networks responsible for stem-like plasticity and cytotoxicity, as revealed by single cell sequencing of the whole transcriptome. Ex vivo, CXCR5+ TIM-3- CD8+ T cells displayed enhanced self-renewal and multipotency compared with more differentiated subsets and were largely polyfunctional. Analysis of inhibitory and costimulatory receptors revealed PD-1, TIGIT, and CD27 as possible targets of immunotherapy. We thus demonstrate a hierarchy of differentiation in the context of T cell exhaustion in human cancer similar to that of chronically infected mice, which is further shown to disappear with disease progression.
Collapse
Affiliation(s)
- Jolanda Brummelman
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Emilia M C Mazza
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Giorgia Alvisi
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Federico S Colombo
- Humanitas Flow Cytometry Core, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Andrea Grilli
- Department of Biological Sciences, University of Modena and Reggio Emilia, Modena, Italy.,PhD Program of Molecular and Translational Medicine, Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate, Italy
| | - Joanna Mikulak
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Domenico Mavilio
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Marco Alloisio
- Division of Thoracic Surgery, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | | | - Egesta Lopci
- Nuclear Medicine Department, Humanitas Clinical and Research Hospital, Milan, Italy
| | - Pierluigi Novellis
- Division of Thoracic Surgery, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Giulia Veronesi
- Division of Thoracic Surgery, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Enrico Lugli
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy .,Humanitas Flow Cytometry Core, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| |
Collapse
|
78
|
Van Acker HH, Campillo-Davo D, Roex G, Versteven M, Smits EL, Van Tendeloo VF. The role of the common gamma-chain family cytokines in γδ T cell-based anti-cancer immunotherapy. Cytokine Growth Factor Rev 2018; 41:54-64. [PMID: 29773448 DOI: 10.1016/j.cytogfr.2018.05.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 05/07/2018] [Accepted: 05/09/2018] [Indexed: 12/28/2022]
Abstract
Cytokines of the common gamma-chain receptor family, comprising interleukin (IL)-2, IL-4, IL-7, IL-9, IL-15 and IL-21, are vital with respect to organizing and sustaining healthy immune cell functions. Supporting the anti-cancer immune response, these cytokines inspire great interest for their use as vaccine adjuvants and cancer immunotherapies. It is against this background that gamma delta (γδ) T cells, as special-force soldiers and natural contributors of the tumor immunosurveillance, also received a lot of attention the last decade. As γδ T cell-based cancer trials are coming of age, this present review focusses on the effects of the different cytokines of the common gamma-chain receptor family on γδ T cells with respect to boosting γδ T cells as a therapeutic target in cancer immunotherapy. This review also gathers data that IL-15 in particular exhibits key features for augmenting the anti-tumor activity of effector killer γδ T cells whilst overcoming the myriad of immune escape mechanisms used by cancer cells.
Collapse
Affiliation(s)
- Heleen H Van Acker
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium.
| | - Diana Campillo-Davo
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Gils Roex
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Maarten Versteven
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Evelien L Smits
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium; Center for Cell Therapy & 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), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| |
Collapse
|
79
|
Khan AB, Carpenter B, Santos e Sousa P, Pospori C, Khorshed R, Griffin J, Velica P, Zech M, Ghorashian S, Forrest C, Thomas S, Gonzalez Anton S, Ahmadi M, Holler A, Flutter B, Ramirez-Ortiz Z, Means TK, Bennett CL, Stauss H, Morris E, Lo Celso C, Chakraverty R. Redirection to the bone marrow improves T cell persistence and antitumor functions. J Clin Invest 2018; 128:2010-2024. [PMID: 29485974 PMCID: PMC5919805 DOI: 10.1172/jci97454] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 02/20/2018] [Indexed: 12/13/2022] Open
Abstract
A key predictor for the success of gene-modified T cell therapies for cancer is the persistence of transferred cells in the patient. The propensity of less differentiated memory T cells to expand and survive efficiently has therefore made them attractive candidates for clinical application. We hypothesized that redirecting T cells to specialized niches in the BM that support memory differentiation would confer increased therapeutic efficacy. We show that overexpression of chemokine receptor CXCR4 in CD8+ T cells (TCXCR4) enhanced their migration toward vascular-associated CXCL12+ cells in the BM and increased their local engraftment. Increased access of TCXCR4 to the BM microenvironment induced IL-15-dependent homeostatic expansion and promoted the differentiation of memory precursor-like cells with low expression of programmed death-1, resistance to apoptosis, and a heightened capacity to generate polyfunctional cytokine-producing effector cells. Following transfer to lymphoma-bearing mice, TCXCR4 showed a greater capacity for effector expansion and better tumor protection, the latter being independent of changes in trafficking to the tumor bed or local out-competition of regulatory T cells. Thus, redirected homing of T cells to the BM confers increased memory differentiation and antitumor immunity, suggesting an innovative solution to increase the persistence and functions of therapeutic T cells.
Collapse
Affiliation(s)
- Anjum B. Khan
- University College London (UCL) Cancer Institute, London, United Kingdom
- UCL Institute of Immunity and Transplantation, London, United Kingdom
| | - Ben Carpenter
- University College London (UCL) Cancer Institute, London, United Kingdom
- UCL Institute of Immunity and Transplantation, London, United Kingdom
| | - Pedro Santos e Sousa
- University College London (UCL) Cancer Institute, London, United Kingdom
- UCL Institute of Immunity and Transplantation, London, United Kingdom
| | - Constandina Pospori
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Reema Khorshed
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - James Griffin
- University College London (UCL) Cancer Institute, London, United Kingdom
- UCL Institute of Immunity and Transplantation, London, United Kingdom
| | - Pedro Velica
- University College London (UCL) Cancer Institute, London, United Kingdom
- UCL Institute of Immunity and Transplantation, London, United Kingdom
| | - Mathias Zech
- UCL Institute of Immunity and Transplantation, London, United Kingdom
| | - Sara Ghorashian
- UCL Institute of Immunity and Transplantation, London, United Kingdom
| | - Calum Forrest
- University College London (UCL) Cancer Institute, London, United Kingdom
| | - Sharyn Thomas
- UCL Institute of Immunity and Transplantation, London, United Kingdom
| | - Sara Gonzalez Anton
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Maryam Ahmadi
- UCL Institute of Immunity and Transplantation, London, United Kingdom
| | - Angelika Holler
- UCL Institute of Immunity and Transplantation, London, United Kingdom
| | - Barry Flutter
- University College London (UCL) Cancer Institute, London, United Kingdom
- UCL Institute of Immunity and Transplantation, London, United Kingdom
| | - Zaida Ramirez-Ortiz
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Terry K. Means
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Clare L. Bennett
- University College London (UCL) Cancer Institute, London, United Kingdom
- UCL Institute of Immunity and Transplantation, London, United Kingdom
| | - Hans Stauss
- UCL Institute of Immunity and Transplantation, London, United Kingdom
| | - Emma Morris
- UCL Institute of Immunity and Transplantation, London, United Kingdom
| | - Cristina Lo Celso
- Department of Life Sciences, Imperial College London, London, United Kingdom
- The Francis Crick Institute, London, United Kingdom
| | - Ronjon Chakraverty
- University College London (UCL) Cancer Institute, London, United Kingdom
- UCL Institute of Immunity and Transplantation, London, United Kingdom
| |
Collapse
|
80
|
Kim G, Hwang H, Jo Y, Lee B, Lee YH, Kim CH, Hong C. Soluble γc receptor attenuates anti-tumor responses of CD8 + T cells in T cell immunotherapy. Int J Cancer 2018; 143:1212-1223. [PMID: 29577276 DOI: 10.1002/ijc.31402] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 02/23/2018] [Accepted: 03/16/2018] [Indexed: 12/22/2022]
Abstract
Previous studies have shown that soluble common γ-chain (sγc) modulates CD4+ T cell immunity with antagonistic functions in γc cytokine signaling. However, the role of sγc in functional properties of effector CD8+ T cells has not been fully defined. In this study, we report a new mechanism by which the anti-tumor activity of mouse CD8+ T cells is suppressed in sγc of their own producing. While sγc significantly inhibits cytotoxicity of CD8+ T cells, blocking sγc production by genetic modification leads to potentiated effector function of CD8+ T cells, establishing persistent CD8+ T cells. This is due to the modulation of IL-2 and IL-15 signaling, which is required for expansion and survival of CD8+ T cells as well as for optimal cytotoxic activity. More efficient management of tumor growth was achieved by an adoptive transfer of sγc-deficient CD8+ T cells than that of wild-type or sγc-overexpressing CD8+ T cells. Blocking of IL-2 and IL-15 signaling by sγc attenuates the capacity of CD8+ T cells to mount an optimal response to the tumor, with both quantitative and qualitative effects on antigen-specific CD8+ T cells. These results could have a critical implication for the generation and survival of optimal effector T cells for adoptive immunotherapy of cancer.
Collapse
Affiliation(s)
- Geona Kim
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, South Korea
| | - Hyunju Hwang
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, South Korea
| | - Yuna Jo
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, South Korea
| | - Byunghyuk Lee
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, South Korea
| | - Young-Ho Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Chan Hyuk Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Changwan Hong
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, South Korea
| |
Collapse
|
81
|
Natural Killer Cells from Malignant Pleural Effusion Are Endowed with a Decidual-Like Proangiogenic Polarization. J Immunol Res 2018; 2018:2438598. [PMID: 29713652 PMCID: PMC5896269 DOI: 10.1155/2018/2438598] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 12/19/2017] [Indexed: 12/28/2022] Open
Abstract
Natural killer (NK) cells are crucial in tumor recognition and eradication, but their activity is impaired in cancer patients, becoming poorly cytotoxic. A particular type of NK cells, from the decidua, has low cytotoxicity and shows proangiogenic functions. We investigated whether NK cells from peripheral blood (PB) and pleural effusions of patients develop decidual-like NK phenotype and whether exposure to IL-2 can restore their killing ability in the presence of pleural fluids. NK cells from pleural effusion of patients with inflammatory conditions (iPE, n = 18), primary tumor (ptPE, n = 18), and metastatic tumor (tmPE, n = 27) acquired the CD56brightCD16− phenotype. NK cells from both ptPE and tmPE showed increased expression for the CD49a and CD69 decidual-like (dNK) markers and decreased levels of the CD57 maturation marker. NK from all the PE analyzed showed impaired degranulation capability and reduced perforin release. PE-NK cells efficiently responded to IL-2 stimulation in vitro. Addition of TGFβ or cell-free pleural fluid to IL-2 in the culture medium abrogated NK cell CD107a and IFNγ expression even in healthy donors (n = 14) NK. We found that tmPE-NK cells produce VEGF and support the formation of capillary-like structures in endothelial cells. Our results suggest that the PE tumor microenvironment can shape NK cell polarization towards a low cytotoxic, decidual-like, highly proangiogenic phenotype and that IL-2 treatment is not sufficient to limit this process.
Collapse
|
82
|
Rosenberg J, Huang J. CD8 + T Cells and NK Cells: Parallel and Complementary Soldiers of Immunotherapy. Curr Opin Chem Eng 2018; 19:9-20. [PMID: 29623254 PMCID: PMC5880541 DOI: 10.1016/j.coche.2017.11.006] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
CD8+ T cells and NK cells are both cytotoxic effector cells of the immune system, but the recognition, specificity, sensitivity, and memory mechanisms are drastically different. While many of these topics have been extensively studied in CD8+ T cells, very little is known about NK cells. Current cancer immunotherapies mainly focus on CD8+ T cells, but have many issues of toxicity and efficacy. Given the heterogeneous nature of cancer, personalized cancer immunotherapy that integrates the power of both CD8+ T cells in adaptive immunity and NK cells in innate immunity might be the future direction, along with precision targeting and effective delivery of tumor-specific, memory CD8+ T cells and NK cells.
Collapse
Affiliation(s)
- Jillian Rosenberg
- Committee on Cancer Biology, The University of Chicago, IL 60637, USA
| | - Jun Huang
- Committee on Cancer Biology, The University of Chicago, IL 60637, USA
- Institute for Molecular Engineering, The University of Chicago, IL 60637, USA
| |
Collapse
|
83
|
Felices M, Lenvik AJ, McElmurry R, Chu S, Hinderlie P, Bendzick L, Geller MA, Tolar J, Blazar BR, Miller JS. Continuous treatment with IL-15 exhausts human NK cells via a metabolic defect. JCI Insight 2018; 3:96219. [PMID: 29415897 PMCID: PMC5821201 DOI: 10.1172/jci.insight.96219] [Citation(s) in RCA: 183] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 01/04/2018] [Indexed: 12/17/2022] Open
Abstract
NK cell-based immunotherapies have been gaining traction in the clinic for treatment of cancer. IL-15 is currently being used in number of clinical trials to improve NK cell expansion and function. The objective of this study is to evaluate the effect of repetitive IL-15 exposure on NK cells. An in vitro model in which human NK cells are continuously (on on on) or intermittently (on off on) treated with IL-15 was used to explore this question. After treatment, cells were evaluated for proliferation, survival, cell cycle gene expression, function, and metabolic processes. Our data indicate that continuous treatment of NK cells with IL-15 resulted in decreased viability and a cell cycle arrest gene expression pattern. This was associated with diminished signaling, decreased function both in vitro and in vivo, and reduced tumor control. NK cells continuously treated with IL-15 also displayed a reduced mitochondrial respiration profile when compared with NK cells treated intermittently with IL-15. This profile was characterized by a decrease in the spare respiratory capacity that was dependent on fatty acid oxidation (FAO). Limiting the strength of IL-15 signaling via utilization of an mTOR inhibitor rescued NK cell functionality in the group continuously treated with IL-15. The findings presented here show that human NK cells continuously treated with IL-15 undergo a process consistent with exhaustion that is accompanied by a reduction in FAO. These findings should inform IL-15-dosing strategies in NK cell cancer immunotherapeutic settings.
Collapse
Affiliation(s)
- Martin Felices
- Department of Medicine, Division of Hematology, Oncology, and Transplantation
| | - Alexander J. Lenvik
- Department of Medicine, Division of Hematology, Oncology, and Transplantation
| | | | - Sami Chu
- Department of Medicine, Division of Hematology, Oncology, and Transplantation
| | - Peter Hinderlie
- Department of Medicine, Division of Hematology, Oncology, and Transplantation
| | - Laura Bendzick
- Department of Obstetrics, Gynecology and Women’s Health, Division of Gynecologic Oncology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Melissa A. Geller
- Department of Obstetrics, Gynecology and Women’s Health, Division of Gynecologic Oncology, University of Minnesota, Minneapolis, Minnesota, USA
| | | | | | - Jeffrey S. Miller
- Department of Medicine, Division of Hematology, Oncology, and Transplantation
| |
Collapse
|
84
|
Zhang L, Romero P. Metabolic Control of CD8+ T Cell Fate Decisions and Antitumor Immunity. Trends Mol Med 2018; 24:30-48. [DOI: 10.1016/j.molmed.2017.11.005] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 11/13/2017] [Accepted: 11/17/2017] [Indexed: 01/20/2023]
|
85
|
de Moura NA, Caetano BFR, de Moraes LN, Carvalho RF, Rodrigues MAM, Barbisan LF. Enhancement of colon carcinogenesis by the combination of indole-3 carbinol and synbiotics in hemin-fed rats. Food Chem Toxicol 2017; 112:11-18. [PMID: 29269057 DOI: 10.1016/j.fct.2017.12.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 12/08/2017] [Accepted: 12/15/2017] [Indexed: 02/07/2023]
Abstract
The risk of developing colorectal cancer (CRC) could be associated with red and processed meat intake. Experimental data supports that hemin iron, found abundantly in red meat, promotes CRC in mice and rats, while indole-3 carbinol (I3C) and synbiotics (syn) exert anti-carcinogenic activities in most studies of colon carcinogenesis. This study aimed to investigate the modifying effects of I3C and syn (inulin + Bifidobacterium lactis), given separately or together, on dimethylhidrazine (DMH)-induced colon carcinogenesis in hemin-fed rats. All animals were given four subcutaneous DMH injections and then, two weeks after carcinogen exposure, they began a basal diet containing hemin, hemin + I3C, hemin + syn, or hemin + I3C + syn for 23 weeks. The combination of I3C + syn significantly increased fecal water genotoxicity, tumor volume and invasiveness when compared to the hemin-fed control group. The groups fed I3C or syn alone had a significant reduction in the number of preneoplastic aberrant crypt foci (ACF) lesions compared to the hemin-fed group. Dietary I3C also reduced fecal water genotoxicity. Gene expression analysis of colorectal tumors demonstrated that the combination of dietary I3C + syn increased transcript levels for Raf1 and decreased tumor progression and invasiveness related to the genes Cdh1 and Appl1. This analysis also revealed that the Tnf and Cdh1 genes were significantly up- and down-regulated, respectively, in tumors of rats that received I3C, in comparison with the hemin-fed group. These findings reveal that the joint administration of I3C and syn enhanced the development of colon tumors induced by DMH in hemin-fed rats, while they potentially reduced ACF development when given alone.
Collapse
Affiliation(s)
- Nelci A de Moura
- Department of Morphology, Institute of Biosciences of Botucatu, Sao Paulo State University (UNESP), Botucatu, SP 18618-689, Brazil
| | - Brunno F R Caetano
- Department of Morphology, Institute of Biosciences of Botucatu, Sao Paulo State University (UNESP), Botucatu, SP 18618-689, Brazil
| | - Leonardo N de Moraes
- Department of Morphology, Institute of Biosciences of Botucatu, Sao Paulo State University (UNESP), Botucatu, SP 18618-689, Brazil
| | - Robson F Carvalho
- Department of Morphology, Institute of Biosciences of Botucatu, Sao Paulo State University (UNESP), Botucatu, SP 18618-689, Brazil
| | - Maria A M Rodrigues
- Department of Pathology, School of Medicine, Sao Paulo State University (UNESP), Botucatu, SP 18610-307, Brazil
| | - Luis F Barbisan
- Department of Morphology, Institute of Biosciences of Botucatu, Sao Paulo State University (UNESP), Botucatu, SP 18618-689, Brazil.
| |
Collapse
|
86
|
Groner B, von Manstein V. Jak Stat signaling and cancer: Opportunities, benefits and side effects of targeted inhibition. Mol Cell Endocrinol 2017; 451:1-14. [PMID: 28576744 DOI: 10.1016/j.mce.2017.05.033] [Citation(s) in RCA: 203] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Accepted: 05/27/2017] [Indexed: 02/06/2023]
Abstract
The effects of Jak Stat signaling and the persistent activation of Stat3 and Stat5 on tumor cell survival, proliferation and invasion have made the Jak Stat pathway a favorite target for drug development and cancer therapy. This notion was strengthened when additional biological functions of Stat signaling in cancer and their roles in the regulation of cytokine dependent inflammation and immunity in the tumor microenvironment were discovered. Stats act not only as transcriptional inducers, but affect gene expression via epigenetic modifications, induce epithelial mesenchymal transition, generate a pro-tumorigenic microenvironment, promote cancer stem cell self-renewal and differentiation, and help to establish the pre-metastatic niche formation. The effects of Jak Stat inhibition on the suppression of pro-inflammatory responses appears most promising and could become a strategy in the prevention of tumor progression. The direct and mediated mechanisms of Jak Stat signaling in and on tumors cells, the interactions with other signaling pathways and transcription factors and the targeting of the functionally crucial secondary modifications of Stat molecules suggest novel approaches to the future development of Jak Stat based cancer therapeutics.
Collapse
Affiliation(s)
- Bernd Groner
- Georg Speyer Haus, Institute for Tumor Biology and Experimental Therapy, Paul Ehrlich Str. 42, D-60596 Frankfurt am Main, Germany.
| | - Viktoria von Manstein
- Georg Speyer Haus, Institute for Tumor Biology and Experimental Therapy, Paul Ehrlich Str. 42, D-60596 Frankfurt am Main, Germany
| |
Collapse
|
87
|
Lugli E, Hudspeth K, Roberto A, Mavilio D. Tissue-resident and memory properties of human T-cell and NK-cell subsets. Eur J Immunol 2017; 46:1809-17. [PMID: 27431095 DOI: 10.1002/eji.201545702] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 06/20/2016] [Accepted: 07/12/2016] [Indexed: 11/11/2022]
Abstract
Efficient immune responses to invading pathogens are the result of the complex but coordinated synergy between a variety of cell types from both the innate and adaptive arms of the immune system. While adaptive and innate immune responses are highly complementary, some cells types within these two systems perform similar functions, underscoring the need for redundancy and increased flexibility. In this review, we will discuss the striking shared features of immunological memory and tissue residency recently discovered between T cells, a component of the adaptive immune system, and natural killer (NK) cells, members generally assigned to the innate compartment. Specifically, we will focus on the T-cell and NK-cell diversity at the single-cell level, on the discrete function of specific subsets, and on their anatomical location. Finally, we will discuss the implication of such diversity in the generation of long-term memory.
Collapse
Affiliation(s)
- Enrico Lugli
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Kelly Hudspeth
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Alessandra Roberto
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Domenico Mavilio
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy.,Department of Medical Biotechnologies and Translational Medicine (BioMeTra), University of Milan, Milan, Italy
| |
Collapse
|
88
|
Lee JH, Tak WY, Lee Y, Heo MK, Song JS, Kim HY, Park SY, Bae SH, Lee JH, Heo J, Kim KH, Bae YS, Kim YJ. Adjuvant immunotherapy with autologous dendritic cells for hepatocellular carcinoma, randomized phase II study. Oncoimmunology 2017; 6:e1328335. [PMID: 28811965 DOI: 10.1080/2162402x.2017.1328335] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 05/04/2017] [Accepted: 05/05/2017] [Indexed: 02/06/2023] Open
Abstract
Our previous phase I/IIA study showed that autologous dendritic cells (DCs) pulsed with tumor-associated antigens are well tolerated in patients with hepatocellular carcinoma (HCC). In this randomized, multicenter, open-label, phase II trial, we investigated the efficacy and safety of this DC-based adjuvant immunotherapy with 156 patients, who treated for HCC with no evidence of residual tumor after standard treatment modalities. Patients were randomly assigned to immunotherapy (n = 77; injection of 3 × 107 DC cells, six times over 14 weeks) or control (n = 79; no treatment). The primary end point was recurrence-free survival (RFS), and the secondary endpoints were immune response and safety. The RFS between the immunotherapy and control groups was not significantly different (hazard ratio [HR], 0.97; 95% confidence interval [CI], 0.60-1.56; p = 0.90). However, post-hoc subgroup analyses revealed that DC immunotherapy significantly reduced the risk of tumor recurrence of non-radiofrequency ablation (non-RFA) group patients (n = 83, HR, 0.49; 95% CI, 0.26-0.94; p = 0.03), whereas unexpectedly increased the risk of recurrence in RFA group (n = 61, p = 0.01). Tumor-specific immune responses were significantly enhanced (both p < 0.01) in the immunotherapy group. Baseline serum interleukin (IL)-15 was statistically correlated with RFS prolongation (HR, 0.16; 95% CI, 0.03-1.58; p = 0.001) within the immunotherapy groups. Overall adverse events were more frequent in the immunotherapy group (p < 0.001) but were mainly mild to moderate in severity. In conclusion, adjuvant immunotherapy with DC vaccine reduces the risk of tumor recurrence in HCC patients who underwent standard treatment modalities other than RFA. Baseline IL-15 might be a candidate biomarker for DC-based HCC immunotherapy.
Collapse
Affiliation(s)
- Jeong-Hoon Lee
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Won Young Tak
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Korea
| | - Yoon Lee
- Department of Biological Science, Sungkyunkwan University, Suwon, Korea.,JW CreaGene Research Institute, JW CreaGene Inc., Seongnam-si, Gyeonggi-do, Korea
| | - Min-Kyu Heo
- JW CreaGene Research Institute, JW CreaGene Inc., Seongnam-si, Gyeonggi-do, Korea
| | - Jae-Sung Song
- JW CreaGene Research Institute, JW CreaGene Inc., Seongnam-si, Gyeonggi-do, Korea
| | - Hak-Yeop Kim
- JW CreaGene Research Institute, JW CreaGene Inc., Seongnam-si, Gyeonggi-do, Korea
| | - Soo Young Park
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Korea
| | - Si Hyun Bae
- The Catholic University Liver Research Center, The Catholic University of Korea, Seoul, Korea
| | - Joon Hyeok Lee
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jeong Heo
- Department of Internal Medicine, Pusan National University School of Medicine, Busan, Republic of Korea
| | - Ki-Hwan Kim
- JW CreaGene Research Institute, JW CreaGene Inc., Seongnam-si, Gyeonggi-do, Korea
| | - Yong-Soo Bae
- Department of Biological Science, Sungkyunkwan University, Suwon, Korea.,JW CreaGene Research Institute, JW CreaGene Inc., Seongnam-si, Gyeonggi-do, Korea
| | - Yoon Jun Kim
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
89
|
Riley JL, Montaner LJ. Cell-Mediated Immunity to Target the Persistent Human Immunodeficiency Virus Reservoir. J Infect Dis 2017; 215:S160-S171. [PMID: 28520969 PMCID: PMC5853458 DOI: 10.1093/infdis/jix002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Effective clearance of virally infected cells requires the sequential activity of innate and adaptive immunity effectors. In human immunodeficiency virus (HIV) infection, naturally induced cell-mediated immune responses rarely eradicate infection. However, optimized immune responses could potentially be leveraged in HIV cure efforts if epitope escape and lack of sustained effector memory responses were to be addressed. Here we review leading HIV cure strategies that harness cell-mediated control against HIV in stably suppressed antiretroviral-treated subjects. We focus on strategies that may maximize target recognition and eradication by the sequential activation of a reconstituted immune system, together with delivery of optimal T-cell responses that can eliminate the reservoir and serve as means to maintain control of HIV spread in the absence of antiretroviral therapy (ART). As evidenced by the evolution of ART, we argue that a combination of immune-based strategies will be a superior path to cell-mediated HIV control and eradication. Available data from several human pilot trials already identify target strategies that may maximize antiviral pressure by joining innate and engineered T cell responses toward testing for sustained HIV remission and/or cure.
Collapse
Affiliation(s)
- James L Riley
- Department of Microbiology and Center for Cellular Immunotherapies, University of Pennsylvania, and
| | - Luis J Montaner
- HIV-1 Immunopathogenesis Laboratory, Wistar Institute, Philadelphia, Pennsylvania
| |
Collapse
|
90
|
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: 442] [Impact Index Per Article: 55.3] [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
|
91
|
Felices M, Chu S, Kodal B, Bendzick L, Ryan C, Lenvik AJ, Boylan KLM, Wong HC, Skubitz APN, Miller JS, Geller MA. IL-15 super-agonist (ALT-803) enhances natural killer (NK) cell function against ovarian cancer. Gynecol Oncol 2017; 145:453-461. [PMID: 28236454 DOI: 10.1016/j.ygyno.2017.02.028] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 02/06/2017] [Accepted: 02/15/2017] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Natural killer (NK) cells represent a powerful immunotherapeutic target as they lyse tumors directly, do not require differentiation, and can elicit potent inflammatory responses. The objective of these studies was to use an IL-15 super-agonist complex, ALT-803 (Altor BioScience Corporation), to enhance the function of both normal and ovarian cancer patient derived NK cells by increasing cytotoxicity and cytokine production. METHODS NK cell function from normal donor peripheral blood mononuclear cells (PBMCs) and ovarian cancer patient ascites was assessed using flow cytometry and chromium release assays ±ALT-803 stimulation. To evaluate the ability of ALT-803 to enhance NK cell function in vivo against ovarian cancer, we used a MA148-luc ovarian cancer NOD scid gamma (NSG) xenogeneic mouse model with transferred human NK cells. RESULTS ALT-803 potently enhanced functionality of NK cells against all ovarian cancer cell lines with significant increases seen in CD107a, IFNγ and TNFα expression depending on target cell line. Function was also rescued in NK cells derived from ovarian cancer patient ascites. Finally, only animals treated with intraperitoneal ALT-803 displayed an NK dependent significant decrease in tumor. CONCLUSIONS ALT-803 enhances NK cell cytotoxicity against ovarian cancer in vitro and in vivo and is able to rescue functionality of NK cells derived from ovarian cancer patient ascites. These findings suggest that ALT-803 has the potential to enhance NK cell-based immunotherapeutic approaches for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- M Felices
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, United States.
| | - S Chu
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, United States
| | - B Kodal
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, United States
| | - L Bendzick
- Department of Obstetrics, Gynecology and Women's Health, Division of Gynecologic Oncology, University of Minnesota, Minneapolis, MN, United States
| | - C Ryan
- Department of Obstetrics, Gynecology and Women's Health, Division of Gynecologic Oncology, University of Minnesota, Minneapolis, MN, United States
| | - A J Lenvik
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, United States
| | - K L M Boylan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States
| | - H C Wong
- Altor BioScience Corporation, Miramar, FL, United States
| | - A P N Skubitz
- Department of Obstetrics, Gynecology and Women's Health, Division of Gynecologic Oncology, University of Minnesota, Minneapolis, MN, United States; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States
| | - J S Miller
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, United States
| | - M A Geller
- Department of Obstetrics, Gynecology and Women's Health, Division of Gynecologic Oncology, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
92
|
Nada MH, Wang H, Workalemahu G, Tanaka Y, Morita CT. Enhancing adoptive cancer immunotherapy with Vγ2Vδ2 T cells through pulse zoledronate stimulation. J Immunother Cancer 2017; 5:9. [PMID: 28239463 PMCID: PMC5319075 DOI: 10.1186/s40425-017-0209-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Accepted: 01/06/2017] [Indexed: 01/14/2023] Open
Abstract
Background Human γδ T cells expressing Vγ2Vδ2 T cell receptors monitor foreign- and self-prenyl pyrophosphate metabolites in isoprenoid biosynthesis to mediate immunity to microbes and tumors. Adoptive immunotherapy with Vγ2Vδ2 T cells has been used to treat cancer patients with partial and complete remissions. Most clinical trials and preclinical studies have used continuous zoledronate exposure to expand Vγ2Vδ2 cells where zoledronate is slowly diluted over the course of the culture. Zoledronate inhibits farnesyl diphosphate synthase (FDPS) in monocytes causing isopentenyl pyrophosphate to accumulate that then stimulates Vγ2Vδ2 cells. Because zoledronate inhibition of FDPS is also toxic for T cells, we hypothesized that a short period of exposure would reduce T cell toxicity but still be sufficient for monocytes uptake. Additionally, IL-15 increases the anti-tumor activity of murine αβ T cells in mice but its effect on the in vivo anti-tumor activity of human Vγ2Vδ2 cells has not been assessed. Methods Human Vγ2Vδ2 T cells were expanded by pulse or continuous zoledronate stimulation with IL-2 or IL-15. Expanded Vγ2Vδ2 cells were tested for their expression of effector molecules and killing of tumor cells as well as their in vivo control of human prostate cancer tumors in immunodeficient NSG mice. Results Pulse zoledronate stimulation with either IL-2 or IL-15 resulted in more uniform expansion of Vγ2Vδ2 cells with higher purity and cell numbers as compared with continuous exposure. The Vγ2Vδ2 cells had higher levels of CD107a and perforin and increased tumor cytotoxicity. Adoptive immunotherapy with Vγ2Vδ2 cells derived by pulse stimulation controlled human PC-3 prostate cancer tumors in NSG mice significantly better than those derived by continuous stimulation, halting tumor growth. Although pulse zoledronate stimulation with IL-15 preserved early memory subsets, adoptive immunotherapy with IL-15-derived Vγ2Vδ2 cells equally inhibited PC-3 tumor growth as those derived with IL-2. Conclusions Pulse zoledronate stimulation maximizes the purity, quantity, and quality of expanded Vγ2Vδ2 cells for adoptive immunotherapy but there is no advantage to using IL-15 over IL-2 in our humanized mouse model. Pulse zoledronate stimulation is a simple modification to existing protocols that will enhance the effectiveness of adoptively transferred Vγ2Vδ2 cells by increasing their numbers and anti-tumor activity. Electronic supplementary material The online version of this article (doi:10.1186/s40425-017-0209-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mohanad H Nada
- Division of Immunology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242 USA.,Department of Veterans Affairs, Iowa City Health Care System, Iowa City, IA 52246 USA.,Interdisciplinary Graduate Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, IA 52242 USA.,Department of Pathology, College of Medicine, Tikrit University, Tikrit, Iraq
| | - Hong Wang
- Division of Immunology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242 USA.,Department of Veterans Affairs, Iowa City Health Care System, Iowa City, IA 52246 USA
| | - Grefachew Workalemahu
- Division of Immunology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242 USA.,Department of Veterans Affairs, Iowa City Health Care System, Iowa City, IA 52246 USA
| | - Yoshimasa Tanaka
- Center for Bioinformatics and Molecular Medicine, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523 Japan
| | - Craig T Morita
- Division of Immunology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242 USA.,Department of Veterans Affairs, Iowa City Health Care System, Iowa City, IA 52246 USA.,Interdisciplinary Graduate Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, IA 52242 USA
| |
Collapse
|
93
|
Croxatto D, Martini S, Chiossone L, Scordamaglia F, Simonassi CF, Moretta L, Mingari MC, Vacca P. IL15 induces a potent antitumor activity in NK cells isolated from malignant pleural effusions and overcomes the inhibitory effect of pleural fluid. Oncoimmunology 2017; 6:e1293210. [PMID: 28507797 DOI: 10.1080/2162402x.2017.1293210] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 02/03/2017] [Accepted: 02/04/2017] [Indexed: 12/14/2022] Open
Abstract
Natural Killer (NK) cells are capable of recognizing and killing cancer cells and play an important role in tumor immunosurveillance. However, tumor-infiltrating NK cells are frequently impaired in their functional capability. A remarkable exception is represented by NK cells isolated from malignant pleural effusions (PE) that are not anergic and, upon IL2-induced activation, efficiently kill tumor cells. Although IL2 is used in various clinical trials, severe side effects may occur in treated patients. In this study, we investigated whether also other clinical-grade cytokines could induce strong cytotoxicity in NK cells isolated from pleural fluid of patients with primary or metastatic tumors of different origins. We show that PE-NK cells, cultured for short-time intervals with IL15, maintain the CD56bright phenotype, a high expression of the main activating receptors, produce cytokines and kill tumor cells in vitro similarly to those treated with IL2. Moreover, IL15-activated PE-NK cells could greatly reduce the growth of established tumors in mice. This in vivo antitumor effect correlated with the ability of IL15-activated PE-NK cells to traffic from periphery to the tumor site. Finally, we show that IL15 can counteract the inhibitory effect of the tumor pleural microenvironment. Our study suggests that IL15-activated NK cells isolated from pleural fluid (otherwise discarded after thoracentesis) may represent a suitable source of effector cells to be used in adoptive immunotherapy of cancer.
Collapse
Affiliation(s)
- D Croxatto
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - S Martini
- IRCCS AOU San Martino-IST, Genoa, Italy
| | - L Chiossone
- Centre d'Immunologie de Marseille-Luminy, Université d'Aix-Marseille UM2, Inserm, U1104, CNRS UMR7280, Marseille, France
| | | | - C F Simonassi
- Department of Pneumology, AO Villa Scassi, Genoa, Italy
| | - L Moretta
- Department of Immunology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - M C Mingari
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy.,IRCCS AOU San Martino-IST, Genoa, Italy
| | - P Vacca
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy.,IRCCS AOU San Martino-IST, Genoa, Italy
| |
Collapse
|
94
|
Abstract
Immuno-oncology (I/O) research has intensified significantly in recent years due to the breakthrough development and the regulatory approval of several immune checkpoint inhibitors, leading to the rapid expansion of the new discovery of novel I/O therapies, new checkpoint inhibitors and beyond. However, many I/O questions remain unanswered, including why only certain subsets of patients respond to these treatments, who the responders would be, and how to expand patient response (the conversion of non-responders or maximizing response in partial responders). All of these require relevant I/O experimental systems, particularly relevant preclinical animal models. Compared to other oncology drug discovery, e.g. cytotoxic and targeted drugs, a lack of relevant animal models is a major obstacle in I/O drug discovery, and an urgent and unmet need. Despite the obvious importance, and the fact that much I/O research has been performed using many different animal models, there are few comprehensive and introductory reviews on this topic. This article attempts to review the efforts in development of a variety of such models, as well as their applications and limitations for readers new to the field, particularly those in the pharmaceutical industry.
Collapse
Affiliation(s)
- Qi-Xiang Li
- Crown Bioscience Inc., 3375 Scott Blvd, Suite 108, Santa Clara, CA 95054, USA; State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China.
| | - Gerold Feuer
- HuMurine Technologies, Inc., 2700 Stockton Blvd, Rm. 1403, Sacramento, CA 95817, USA
| | - Xuesong Ouyang
- Crown Bioscience Inc., 3375 Scott Blvd, Suite 108, Santa Clara, CA 95054, USA
| | - Xiaoyu An
- Crown Bioscience Inc., 3375 Scott Blvd, Suite 108, Santa Clara, CA 95054, USA; State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| |
Collapse
|
95
|
Fabbi M, Carbotti G, Ferrini S. Dual Roles of IL-27 in Cancer Biology and Immunotherapy. Mediators Inflamm 2017; 2017:3958069. [PMID: 28255204 PMCID: PMC5309407 DOI: 10.1155/2017/3958069] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/23/2016] [Accepted: 01/12/2017] [Indexed: 01/09/2023] Open
Abstract
IL-27 is a pleiotropic two-chain cytokine, composed of EBI3 and IL-27p28 subunits, which is structurally related to both IL-12 and IL-6 cytokine families. IL-27 acts through a heterodimer receptor consisting of IL-27Rα (WSX1) and gp130 chains, which mediate signaling predominantly through STAT1 and STAT3. IL-27 was initially reported as an immune-enhancing cytokine that supports CD4+ T cell proliferation, T helper (Th)1 cell differentiation, and IFN-γ production, acting in concert with IL-12. However, subsequent studies demonstrated that IL-27 displays complex immune-regulatory functions, which may result in either proinflammatory or anti-inflammatory effects in relationship to the biological context and experimental models considered. Several pieces of evidence, obtained in preclinical tumor models, indicated that IL-27 has a potent antitumor activity, related not only to the induction of tumor-specific Th1 and cytotoxic T lymphocyte (CTL) responses but also to direct inhibitory effects on tumor cell proliferation, survival, invasiveness, and angiogenic potential. Nonetheless, given its immune-regulatory functions, the effects of IL-27 on cancer may be dual and protumor effects may also occur. Here, we will summarize IL-27 biological activities and its functional overlaps with the IFNs and discuss its dual role in tumors in the light of potential applications to cancer immunotherapy.
Collapse
Affiliation(s)
- Marina Fabbi
- Laboratory of Biotherapy, IRCCS AOU San Martino-IST, Istituto Nazionale per la Ricerca sul Cancro, 16132 Genoa, Italy
| | - Grazia Carbotti
- Laboratory of Biotherapy, IRCCS AOU San Martino-IST, Istituto Nazionale per la Ricerca sul Cancro, 16132 Genoa, Italy
| | - Silvano Ferrini
- Laboratory of Biotherapy, IRCCS AOU San Martino-IST, Istituto Nazionale per la Ricerca sul Cancro, 16132 Genoa, Italy
| |
Collapse
|
96
|
Pellom ST, Dudimah DF, Thounaojam MC, Uzhachenko RV, Singhal A, Richmond A, Shanker A. Bortezomib augments lymphocyte stimulatory cytokine signaling in the tumor microenvironment to sustain CD8+T cell antitumor function. Oncotarget 2017; 8:8604-8621. [PMID: 28052005 PMCID: PMC5352426 DOI: 10.18632/oncotarget.14365] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 12/07/2016] [Indexed: 12/19/2022] Open
Abstract
Tumor-induced immune tolerance poses a major challenge for therapeutic interventions aimed to manage cancer. We explored approaches to overcome T-cell suppression in murine breast and kidney adenocarcinomas, and lung fibrosarcoma expressing immunogenic antigens. We observed that treatment with a reversible proteasome inhibitor bortezomib (1 mg/kg body weight) in tumor-bearing mice significantly enhanced the expression of lymphocyte-stimulatory cytokines IL-2, IL-12, and IL-15. Notably, bortezomib administration reduced pulmonary nodules of mammary adenocarcinoma 4T1.2 expressing hemagglutinin (HA) model antigen (4T1HA) in mice. Neutralization of IL-12 and IL-15 cytokines with a regimen of blocking antibodies pre- and post-adoptive transfer of low-avidity HA518-526-specific CD8+T-cells following intravenous injection of 4T1HA cells increased the number of pulmonary tumor nodules. This neutralization effect was counteracted by the tumor metastasis-suppressing action of bortezomib treatments. In bortezomib-treated 4T1HA tumor-bearing mice, CD4+T-cells showed increased IL-2 production, CD11c+ dendritic cells showed increased IL-12 and IL-15 production, and HA-specific activated CD8+T-cells showed enhanced expression of IFNγ, granzyme-B and transcription factor eomesodermin. We also noted a trend of increased expression of IL-2, IL-12 and IL-15 receptors as well as increased phosphorylation of STAT5 in tumor-infiltrating CD8+T-cells following bortezomib treatment. Furthermore, bortezomib-treated CD8+T-cells showed increased phosphorylation of mitogen-activated protein kinase p38, and Akt, which was abrogated by phosphatidylinositide 3-kinase (PI3K) inhibitor. These data support the therapeutic potential of bortezomib in conjunction with other immunotherapies to augment the strength of convergent signals from CD8+T-cell signaling molecules including TCR, cytokine receptors and downstream PI3K/Akt/STAT5 pathways to sustain CD8+T-cell effector function in the tumor microenvironment.
Collapse
Affiliation(s)
- Samuel T. Pellom
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, Tennessee, USA
- Department of Microbiology and Immunology, School of Medicine, Meharry Medical College, Nashville, Tennessee, USA
- School of Graduate Studies and Research, Meharry Medical College, Nashville, Tennessee, USA
| | - Duafalia F. Dudimah
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, Tennessee, USA
| | - Menaka C. Thounaojam
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, Tennessee, USA
| | - Roman V. Uzhachenko
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, Tennessee, USA
| | - Ashutosh Singhal
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, Tennessee, USA
| | - Ann Richmond
- Tennessee Valley Healthcare System, Nashville, Tennessee, USA
- Department of Veterans Affairs, Nashville, Tennessee, USA
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Host-Tumor Interactions Research Program, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Center for Translational and Clinical Immunology, Vanderbilt University, Nashville, Tennessee, USA
| | - Anil Shanker
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, Tennessee, USA
- School of Graduate Studies and Research, Meharry Medical College, Nashville, Tennessee, USA
- Host-Tumor Interactions Research Program, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Center for Translational and Clinical Immunology, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
97
|
Ng S, Deng J, Chinnadurai R, Yuan S, Pennati A, Galipeau J. Stimulation of Natural Killer Cell-Mediated Tumor Immunity by an IL15/TGFβ-Neutralizing Fusion Protein. Cancer Res 2016; 76:5683-5695. [PMID: 27488533 DOI: 10.1158/0008-5472.can-16-0386] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 06/19/2016] [Indexed: 11/16/2022]
Abstract
The clinical efficacy of immune cytokines used for cancer therapy is hampered by elements of the immunosuppressive tumor microenvironment such as TGFβ. Here we demonstrate that FIST15, a recombinant chimeric protein composed of the T-cell-stimulatory cytokine IL15, the sushi domain of IL15Rα and a TGFβ ligand trap, can overcome immunosuppressive TGFβ to effectively stimulate the proliferation and activation of natural killer (NK) and CD8+ T cells with potent antitumor properties. FIST15-treated NK and CD8+ T cells produced more IFNγ and TNFα compared with treatment with IL15 and a commercially available TGFβ receptor-Fc fusion protein (sTβRII) in the presence of TGFβ. Murine B16 melanoma cells, which overproduce TGFβ, were lysed by FIST15-treated NK cells in vitro at doses approximately 10-fold lower than NK cells treated with IL15 and sTβRII. Melanoma cells transduced to express FIST15 failed to establish tumors in vivo in immunocompetent murine hosts and could only form tumors in beige mice lacking NK cells. Mice injected with the same cells were also protected from subsequent challenge by unmodified B16 melanoma cells. Finally, mice with pre-established B16 melanoma tumors responded to FIST15 treatment more strongly compared with tumors treated with control cytokines. Taken together, our results offer a preclinical proof of concept for the use of FIST15 as a new class of biological therapeutics that can coordinately neutralize the effects of immunosuppressive TGFβ in the tumor microenvironment while empowering tumor immunity. Cancer Res; 76(19); 5683-95. ©2016 AACR.
Collapse
Affiliation(s)
- Spencer Ng
- Department of Hematology and Medical Oncology, School of Medicine, Emory University, Atlanta, Georgia. Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Jiusheng Deng
- Department of Hematology and Medical Oncology, School of Medicine, Emory University, Atlanta, Georgia. Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Raghavan Chinnadurai
- Department of Hematology and Medical Oncology, School of Medicine, Emory University, Atlanta, Georgia. Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Shala Yuan
- Department of Hematology and Medical Oncology, School of Medicine, Emory University, Atlanta, Georgia. Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Andrea Pennati
- Department of Hematology and Medical Oncology, School of Medicine, Emory University, Atlanta, Georgia. Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Jacques Galipeau
- Department of Hematology and Medical Oncology, School of Medicine, Emory University, Atlanta, Georgia. Winship Cancer Institute, Emory University, Atlanta, Georgia. Department of Pediatrics, School of Medicine, Emory University, Atlanta, Georgia.
| |
Collapse
|
98
|
Tsukahara T, Hirohashi Y, Kanaseki T, Nakatsugawa M, Kubo T, Sato N, Torigoe T. Peptide vaccination therapy: Towards the next generation. Pathol Int 2016; 66:547-553. [PMID: 27435148 DOI: 10.1111/pin.12438] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 06/14/2016] [Accepted: 06/24/2016] [Indexed: 12/24/2022]
Affiliation(s)
- Tomohide Tsukahara
- Department of PathologySapporo Medical University School of Medicine South‐1 West‐17, Chuo‐ku Sapporo Japan
| | - Yoshihiko Hirohashi
- Department of PathologySapporo Medical University School of Medicine South‐1 West‐17, Chuo‐ku Sapporo Japan
| | - Takayuki Kanaseki
- Department of PathologySapporo Medical University School of Medicine South‐1 West‐17, Chuo‐ku Sapporo Japan
| | - Munehide Nakatsugawa
- Department of PathologySapporo Medical University School of Medicine South‐1 West‐17, Chuo‐ku Sapporo Japan
| | - Terufumi Kubo
- Department of PathologySapporo Medical University School of Medicine South‐1 West‐17, Chuo‐ku Sapporo Japan
| | - Noriyuki Sato
- Department of PathologySapporo Medical University School of Medicine South‐1 West‐17, Chuo‐ku Sapporo Japan
| | - Toshihiko Torigoe
- Department of PathologySapporo Medical University School of Medicine South‐1 West‐17, Chuo‐ku Sapporo Japan
| |
Collapse
|