51
|
Modak S, Le Luduec JB, Cheung IY, Goldman DA, Ostrovnaya I, Doubrovina E, Basu E, Kushner BH, Kramer K, Roberts SS, O'Reilly RJ, Cheung NKV, Hsu KC. Adoptive immunotherapy with haploidentical natural killer cells and Anti-GD2 monoclonal antibody m3F8 for resistant neuroblastoma: Results of a phase I study. Oncoimmunology 2018; 7:e1461305. [PMID: 30221057 DOI: 10.1080/2162402x.2018.1461305] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 03/27/2018] [Accepted: 03/30/2018] [Indexed: 01/30/2023] Open
Abstract
Natural killer (NK) cell-mediated antibody-dependent toxicity is a potent mechanism of action of the anti-GD2 murine monoclonal antibody 3F8 (m3F8). Killer immunoglobulin-like receptor (KIR) and HLA genotypes modulate NK activity and are key prognostic markers in m3F8-treated patients with neuroblastoma. Endogenous NK-cells are suppressed in the setting of high tumor burden and chemotherapy. Allogeneic NK-cells however, demonstrate potent anti-neuroblastoma activity. We report on the results of a phase I clinical trial of haploidentical NK-cells plus m3F8 administered to patients with high-risk neuroblastoma after conditioning chemotherapy. The primary objective was to determine the maximum tolerated NK-cell dose (MTD). Secondary objectives included assessing anti-neuroblastoma activity and its relationship to donor-recipient KIR/HLA genotypes, NK function, and donor NK chimerism. Patients received a lymphodepleting regimen prior to infusion of haploidentical CD3-CD56+ NK-cells, followed by m3F8. Overall and progression free survival (PFS) were assessed from the time of first NK-cell dose. Univariate Cox regression assessed relationship between dose and outcomes. Thirty-five patients received NK-cells at one of five dose levels ranging from <1×106 to 50×106 CD3-CD56+cells/kg. One patient experienced grade 3 hypertension and grade 4 pneumonitis. MTD was not reached. Ten patients (29%) had complete or partial response; 17 (47%) had no response; and eight (23%) had progressive disease. No relationship was found between response and KIR/HLA genotype or between response and FcγRIII receptor polymorphisms. Patients receiving >10×106 CD56+cells/kg had improved PFS (HR: 0.36, 95%CI: 0.15-0.87, p = 0.022). Patient NK-cells displayed high NKG2A expression, leading to inhibition by HLA-E-expressing neuroblastoma cells. Adoptive NK-cell therapy in combination with m3F8 is safe and has anti-neuroblastoma activity at higher cell doses.
Collapse
Affiliation(s)
- Shakeel Modak
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Irene Y Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Debra A Goldman
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Irina Ostrovnaya
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Ellen Basu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Brian H Kushner
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kim Kramer
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Stephen S Roberts
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Richard J O'Reilly
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY.,Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, NY
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Katharine C Hsu
- Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, NY.,Weill Cornell Medical College, New York, NY.,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
52
|
Nakagawara A, Li Y, Izumi H, Muramori K, Inada H, Nishi M. Neuroblastoma. Jpn J Clin Oncol 2018; 48:214-241. [PMID: 29378002 DOI: 10.1093/jjco/hyx176] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Indexed: 02/07/2023] Open
Abstract
Neuroblastoma is one of the most common solid tumors in children and has a diverse clinical behavior that largely depends on the tumor biology. Neuroblastoma exhibits unique features, such as early age of onset, high frequency of metastatic disease at diagnosis in patients over 1 year of age and the tendency for spontaneous regression of tumors in infants. The high-risk tumors frequently have amplification of the MYCN oncogene as well as segmental chromosome alterations with poor survival. Recent advanced genomic sequencing technology has revealed that mutation of ALK, which is present in ~10% of primary tumors, often causes familial neuroblastoma with germline mutation. However, the frequency of gene mutations is relatively small and other aberrations, such as epigenetic abnormalities, have also been proposed. The risk-stratified therapy was introduced by the Japan Neuroblastoma Study Group (JNBSG), which is now moving to the Neuroblastoma Committee of Japan Children's Cancer Group (JCCG). Several clinical studies have facilitated the reduction of therapy for children with low-risk neuroblastoma disease and the significant improvement of cure rates for patients with intermediate-risk as well as high-risk disease. Therapy for patients with high-risk disease includes intensive induction chemotherapy and myeloablative chemotherapy, followed by the treatment of minimal residual disease using differentiation therapy and immunotherapy. The JCCG aims for better cures and long-term quality of life for children with cancer by facilitating new approaches targeting novel driver proteins, genetic pathways and the tumor microenvironment.
Collapse
Affiliation(s)
| | - Yuanyuan Li
- Laboratory of Molecular Biology, Life Science Research Institute, Saga Medical Center Koseikan
| | - Hideki Izumi
- Laboratory of Molecular Biology, Life Science Research Institute, Saga Medical Center Koseikan
| | | | - Hiroko Inada
- Department of Pediatrics, Saga Medical Center Koseikan
| | - Masanori Nishi
- Department of Pediatrics, Saga University, Saga 849-8501, Japan
| |
Collapse
|
53
|
Abstract
Neuroblastoma (NB) is the most common solid childhood tumor outside the brain and causes 15% of childhood cancer-related mortality. The main drivers of NB formation are neural crest cell-derived sympathoadrenal cells that undergo abnormal genetic arrangements. Moreover, NB is a complex disease that has high heterogeneity and is therefore difficult to target for successful therapy. Thus, a better understanding of NB development helps to improve treatment and increase the survival rate. One of the major causes of sporadic NB is known to be MYCN amplification and mutations in ALK (anaplastic lymphoma kinase) are responsible for familial NB. Many other genetic abnormalities can be found; however, they are not considered as driver mutations, rather they support tumor aggressiveness. Tumor cell elimination via cell death is widely accepted as a successful technique. Therefore, in this review, we provide a thorough overview of how different modes of cell death and treatment strategies, such as immunotherapy or spontaneous regression, are or can be applied for NB elimination. In addition, several currently used and innovative approaches and their suitability for clinical testing and usage will be discussed. Moreover, significant attention will be given to combined therapies that show more effective results with fewer side effects than drugs targeting only one specific protein or pathway.
Collapse
|
54
|
The role of interleukin-2, all-trans retinoic acid, and natural killer cells: surveillance mechanisms in anti-GD2 antibody therapy in neuroblastoma. Cancer Immunol Immunother 2018; 67:615-626. [PMID: 29327110 DOI: 10.1007/s00262-017-2108-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 12/14/2017] [Indexed: 02/05/2023]
Abstract
Although anti-disialoganglioside (GD2) antibodies are successfully used for neuroblastoma therapy, a third of patients with neuroblastoma experience treatment failure or serious toxicity. Various strategies have been employed in the clinic to improve antibody-dependent cell-mediated cytotoxicity (ADCC), such as the addition of interleukin (IL)-2 to enhance natural killer (NK) cell function, adoptive transfer of allogeneic NK cells to exploit immune surveillance, and retinoid-induced differentiation therapy. Nevertheless, these mechanisms are not fully understood. We developed a quantitative assay to test ADCC induced by the anti-GD2 antibody Hu14.18K322A in nine neuroblastoma cell lines and dissociated cells from orthotopic patient-derived xenografts (O-PDXs) in culture. IL-2 improved ADCC against neuroblastoma cells, and differentiation with all-trans retinoic acid stabilized GD2 expression on tumor cells and enhanced ADCC as well. Degranulation was highest in licensed NK cells that expressed CD158b (P < 0.001) and harbored a killer-cell immunoglobulin-like receptor (KIR) mismatch against the tumor-specific human leukocyte antigen (HLA; P = 0.016). In conclusion, IL-2 is an important component of immunotherapy because it can improve the cytolytic function of NK cells against neuroblastoma cells and could lower the antibody dose required for efficacy, thereby reducing toxicity. The effect of IL-2 may vary among individuals and a biomarker would be useful to predict ADCC following IL-2 activation. Sub-populations of NK cells may have different levels of activity dependent on their licensing status, KIR expression, and HLA-KIR interaction. Better understanding of HLA-KIR interactions and the molecular changes following retinoid-induced differentiation is necessary to delineate their role in ADCC.
Collapse
|
55
|
Cheung NK, Hsu KC. Genotyping Natural Killer Immune Checkpoints to Discover Biomarkers of Response. Clin Cancer Res 2017; 24:3-5. [PMID: 29122934 DOI: 10.1158/1078-0432.ccr-17-2884] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 10/31/2017] [Accepted: 11/07/2017] [Indexed: 11/16/2022]
Abstract
Immune checkpoints have been a focus of immunotherapy in the recent decade. Killer cell immunoglobulin-like receptors (KIR) and their cognate human leukocyte antigen (HLA) class I ligands have evolved as checkpoints to ensure self-tolerance of natural killer cells. Both KIR and HLA genetic profiles are potential biomarkers of immunotherapy outcome. Clin Cancer Res; 24(1); 3-5. ©2017 AACRSee related article by Erbe et al., p. 189.
Collapse
Affiliation(s)
- Nai-Kong Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Katharine C Hsu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
56
|
Erbe AK, Wang W, Carmichael L, Kim K, Mendonça EA, Song Y, Hess D, Reville PK, London WB, Naranjo A, Hank JA, Diccianni MB, Reisfeld RA, Gillies SD, Matthay KK, Cohn SL, Hogarty MD, Maris JM, Park JR, Ozkaynak MF, Gilman AL, Yu AL, Sondel PM. Neuroblastoma Patients' KIR and KIR-Ligand Genotypes Influence Clinical Outcome for Dinutuximab-based Immunotherapy: A Report from the Children's Oncology Group. Clin Cancer Res 2017; 24:189-196. [PMID: 28972044 DOI: 10.1158/1078-0432.ccr-17-1767] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 08/24/2017] [Accepted: 09/22/2017] [Indexed: 11/16/2022]
Abstract
Purpose: In 2010, a Children's Oncology Group (COG) phase III randomized trial for patients with high-risk neuroblastoma (ANBL0032) demonstrated improved event-free survival (EFS) and overall survival (OS) following treatment with an immunotherapy regimen of dinutuximab, GM-CSF, IL2, and isotretinoin compared with treatment with isotretinoin alone. Dinutuximab, a chimeric anti-GD2 monoclonal antibody, acts in part via natural killer (NK) cells. Killer immunoglobulin-like receptors (KIR) on NK cells and their interactions with KIR-ligands can influence NK cell function. We investigated whether KIR/KIR-ligand genotypes were associated with EFS or OS in this trial.Experimental Design: We genotyped patients from COG study ANBL0032 and evaluated the effect of KIR/KIR-ligand genotypes on clinical outcomes. Cox regression models and log-rank tests were used to evaluate associations of EFS and OS with KIR/KIR-ligand genotypes.Results: In this trial, patients with the "all KIR-ligands present" genotype as well as patients with inhibitory KIR2DL2 with its ligand (HLA-C1) together with inhibitory KIR3DL1 with its ligand (HLA-Bw4) were associated with improved outcome if they received immunotherapy. In contrast, for patients with the complementary KIR/KIR-ligand genotypes, clinical outcome was not significantly different for patients who received immunotherapy versus those receiving isotretinoin alone.Conclusions: These data show that administration of immunotherapy is associated with improved outcome for neuroblastoma patients with certain KIR/KIR-ligand genotypes, although this was not seen for patients with other KIR/KIR-ligand genotypes. Further investigation of KIR/KIR-ligand genotypes may clarify their role in cancer immunotherapy and may enable KIR/KIR-ligand genotyping to be used prospectively for identifying patients likely to benefit from certain cancer immunotherapy regimens. Clin Cancer Res; 24(1); 189-96. ©2017 AACRSee related commentary by Cheung and Hsu, p. 3.
Collapse
Affiliation(s)
- Amy K Erbe
- Department of Human Oncology, University of Wisconsin, Madison, Wisconsin
| | - Wei Wang
- Department of Human Oncology, University of Wisconsin, Madison, Wisconsin
| | - Lakeesha Carmichael
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, Wisconsin
| | - KyungMann Kim
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, Wisconsin
| | - Eneida A Mendonça
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, Wisconsin.,Department of Pediatrics, University of Wisconsin, Madison, Wisconsin
| | - Yiqiang Song
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, Wisconsin
| | - Dustin Hess
- Department of Human Oncology, University of Wisconsin, Madison, Wisconsin
| | - Patrick K Reville
- Department of Human Oncology, University of Wisconsin, Madison, Wisconsin
| | - Wendy B London
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, Massachusetts
| | - Arlene Naranjo
- COG Statistics and Data Center, Department of Biostatistics, University of Florida, Gainesville, Florida
| | - Jacquelyn A Hank
- Department of Human Oncology, University of Wisconsin, Madison, Wisconsin
| | - Mitchell B Diccianni
- Department of Pediatrics, Hematology/Oncology and Moores Cancer Center, University of California, San Diego, California
| | | | | | - Katherine K Matthay
- UCSF Benioff Children's Hospital and University of California School of Medicine, San Francisco, California
| | - Susan L Cohn
- Department of Pediatrics, University of Chicago, Chicago, Illinois
| | - Michael D Hogarty
- Children's Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - John M Maris
- Children's Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Julie R Park
- Seattle Children's Hospital, Seattle, Washington.,University of Washington, Seattle, Washington
| | | | | | - Alice L Yu
- Department of Pediatrics, Hematology/Oncology and Moores Cancer Center, University of California, San Diego, California.,Institute of Stem Cell & Translational Cancer Research, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Paul M Sondel
- Department of Human Oncology, University of Wisconsin, Madison, Wisconsin. .,Department of Pediatrics, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
57
|
Abstract
INTRODUCTION Current therapeutic approaches for high-risk neuroblastoma (HR-NB) include high-dose chemotherapy, surgery and radiotherapy; interventions that are associated with long and short-term toxicities. Effective immunotherapy holds particular promise for improving survival and quality of life by reducing exposure to cytotoxic agents. GD2, a surface glycolipid is the most common target for immunotherapy. Areas covered: We review the status of anti-GD2 immunotherapies currently in clinical use for neuroblastomas and novel GD2-targeted strategies in preclinical development. Expert commentary: Anti-GD2 monoclonal antibodies are associated with improved survival in patients in their first remission and are increasingly being used for chemorefractory and relapsed neuroblastoma. As protein engineering technology has become more accessible, newer antibody constructs are being tested. GD2 is also being targeted by natural killer cells and T-cells. Active immunity can be elicited by anti-GD2 vaccines. The rational combination of currently available and soon-to-emerge immunotherapeutic approaches, and their integration into conventional multimodality therapies will require further investigation to optimize their use for HR-NB.
Collapse
Affiliation(s)
- Sameer Sait
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Shakeel I. Modak
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
58
|
Erbe AK, Wang W, Reville PK, Carmichael L, Kim K, Mendonca EA, Song Y, Hank JA, London WB, Naranjo A, Hong F, Hogarty MD, Maris JM, Park JR, Ozkaynak MF, Miller JS, Gilman AL, Kahl B, Yu AL, Sondel PM. HLA-Bw4-I-80 Isoform Differentially Influences Clinical Outcome As Compared to HLA-Bw4-T-80 and HLA-A-Bw4 Isoforms in Rituximab or Dinutuximab-Based Cancer Immunotherapy. Front Immunol 2017; 8:675. [PMID: 28659916 PMCID: PMC5466980 DOI: 10.3389/fimmu.2017.00675] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 05/24/2017] [Indexed: 11/21/2022] Open
Abstract
Killer-cell immunoglobulin-like receptors (KIRs) are a family of glycoproteins expressed primarily on natural killer cells that can regulate their function. Inhibitory KIRs recognize MHC class I molecules (KIR-ligands) as ligands. We have reported associations of KIRs and KIR-ligands for patients in two monoclonal antibody (mAb)-based trials: (1) A Children’s Oncology Group (COG) trial for children with high-risk neuroblastoma randomized to immunotherapy treatment with dinutuximab (anti-GD2 mAb) + GM-CSF + IL-2 + isotretinion or to treatment with isotretinoin alone and (2) An Eastern Cooperative Oncology Group (ECOG) trial for adults with low-tumor burden follicular lymphoma responding to an induction course of rituximab (anti-CD20 mAb) and randomized to treatment with maintenance rituximab or no-maintenance rituximab. In each trial, certain KIR/KIR-ligand genotypes were associated with clinical benefit for patients randomized to immunotherapy treatment (immunotherapy in COG; maintenance rituximab in ECOG) as compared to patients that did not receive the immunotherapy [isotretinoin alone (COG); no-maintenance (ECOG)]. Namely, patients with both KIR3DL1 and its HLA-Bw4 ligand (KIR3DL1+/HLA-Bw4+ genotype) had improved clinical outcomes if randomized to immunotherapy regimens, as compared to patients with the KIR3DL1+/HLA-Bw4+ genotype randomized to the non-immunotherapy regimen. Conversely, patients that did not have the KIR3DL1+/HLA-Bw4+ genotype showed no evidence of a difference in outcome if receiving the immunotherapy vs. no-immunotherapy. For each trial, HLA-Bw4 status was determined by assessing the genotypes of three separate isoforms of HLA-Bw4: (1) HLA-B-Bw4 with threonine at amino acid 80 (B-Bw4-T80); (2) HLA-B-Bw4 with isoleucine at amino acid 80 (HLA-B-Bw4-I80); and (3) HLA-A with a Bw4 epitope (HLA-A-Bw4). Here, we report on associations with clinical outcome for patients with KIR3DL1 and these separate isoforms of HLA-Bw4. Patients randomized to immunotherapy with KIR3DL1+/A-Bw4+ or with KIR3DL1+/B-Bw4-T80+ had better outcome vs. those randomized to no-immunotherapy, whereas for those with KIR3DL1+/B-Bw4-I80+ there was no evidence of a difference based on immunotherapy vs. no-immunotherapy. Additionally, we observed differences within treatment types (either within immunotherapy or no-immunotherapy) that were associated with the genotype status for the different KIR3DL1/HLA-Bw4-isoforms. These studies suggest that specific HLA-Bw4 isoforms may differentially influence response to these mAb-based immunotherapy, further confirming the involvement of KIR-bearing cells in tumor-reactive mAb-based cancer immunotherapy.
Collapse
Affiliation(s)
- Amy K Erbe
- Department of Human Oncology, University of Wisconsin, Madison, WI, United States
| | - Wei Wang
- Department of Human Oncology, University of Wisconsin, Madison, WI, United States
| | - Patrick K Reville
- Department of Human Oncology, University of Wisconsin, Madison, WI, United States
| | - Lakeesha Carmichael
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, WI, United States
| | - KyungMann Kim
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, WI, United States
| | - Eneida A Mendonca
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, WI, United States
| | - Yiqiang Song
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, WI, United States
| | - Jacquelyn A Hank
- Department of Human Oncology, University of Wisconsin, Madison, WI, United States
| | - Wendy B London
- Dana-Farber Cancer Institute/Boston Children's Cancer and Blood Disorder Center, Harvard Medical School, Boston, MA, United States
| | - Arlene Naranjo
- COG Statistics and Data Center, Department of Biostatistics, University of Florida, Gainesville, FL, United States
| | - Fangxin Hong
- Department of Biostatistics, Harvard University, Dana Farber Cancer Institute, Boston, MA, United States
| | - Michael D Hogarty
- Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, PA, United States
| | - John M Maris
- Provenance Biopharmaceuticals, Carlisle, MA, United States
| | - Julie R Park
- Seattle Children's Hospital/University, Seattle, WA, United States.,University of Washington, Seattle, WA, United States
| | - M F Ozkaynak
- New York Medical College, Valhalla, NY, United States
| | - Jeffrey S Miller
- Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | | | - Brad Kahl
- Department of Medicine, Washington University, St. Louis, MO, United States
| | - Alice L Yu
- Department of Pediatrics, Hematology/Oncology, Moores Cancer Center, University of California San Diego, San Diego, CA, United States.,Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Paul M Sondel
- Department of Human Oncology, University of Wisconsin, Madison, WI, United States.,Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
59
|
Guillerey C, Huntington ND, Smyth MJ. Targeting natural killer cells in cancer immunotherapy. Nat Immunol 2017; 17:1025-36. [PMID: 27540992 DOI: 10.1038/ni.3518] [Citation(s) in RCA: 812] [Impact Index Per Article: 101.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 06/22/2016] [Indexed: 12/14/2022]
Abstract
Alteration in the expression of cell-surface proteins is a common consequence of malignant transformation. Natural killer (NK) cells use an array of germline-encoded activating and inhibitory receptors that scan for altered protein-expression patterns, but tumor evasion of detection by the immune system is now recognized as one of the hallmarks of cancer. NK cells display rapid and potent immunity to metastasis or hematological cancers, and major efforts are now being undertaken to fully exploit NK cell anti-tumor properties in the clinic. Diverse approaches encompass the development of large-scale NK cell-expansion protocols for adoptive transfer, the establishment of a microenvironment favorable to NK cell activity, the redirection of NK cell activity against tumor cells and the release of inhibitory signals that limit NK cell function. In this Review we detail recent advances in NK cell-based immunotherapies and discuss the advantages and limitations of these strategies.
Collapse
Affiliation(s)
- Camille Guillerey
- Immunology of Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Australia.,School of Medicine, University of Queensland, Herston, Australia
| | - Nicholas D Huntington
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, Australia
| | - Mark J Smyth
- Immunology of Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Australia.,School of Medicine, University of Queensland, Herston, Australia
| |
Collapse
|
60
|
Bernson E, Hallner A, Sander FE, Wilsson O, Werlenius O, Rydström A, Kiffin R, Brune M, Foà R, Aurelius J, Martner A, Hellstrand K, Thorén FB. Impact of killer-immunoglobulin-like receptor and human leukocyte antigen genotypes on the efficacy of immunotherapy in acute myeloid leukemia. Leukemia 2017; 31:2552-2559. [PMID: 28529313 PMCID: PMC5729331 DOI: 10.1038/leu.2017.151] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 05/03/2017] [Accepted: 05/09/2017] [Indexed: 12/23/2022]
Abstract
Interactions between killer-immunoglobulin-like receptors (KIRs) and their HLA class I ligands are instrumental in natural killer (NK) cell regulation and protect normal tissue from NK cell attack. Human KIR haplotypes comprise genes encoding mainly inhibitory receptors (KIR A) or activating and inhibitory receptors (KIR B). A substantial fraction of humans lack ligands for inhibitory KIRs (iKIRs), that is, a ‘missing ligand’ genotype. KIR B/x and missing ligand genotypes may thus give rise to potentially autoreactive, unlicensed NK cells. Little is known regarding the impact of such genotypes in untransplanted acute myeloid leukemia (AML). For this study, NK cell phenotypes and KIR/HLA genotypes were determined in 81 AML patients who received immunotherapy with histamine dihydrochloride and low-dose IL-2 for relapse prevention (NCT01347996). We observed that presence of unlicensed NK cells impacted favorably on clinical outcome, in particular among patients harboring functional NK cells reflected by high expression of the natural cytotoxicity receptor (NCR) NKp46. Genotype analyses suggested that the clinical benefit of high NCR expression was restricted to patients with a missing ligand genotype and/or a KIR B/x genotype. These data imply that functional NK cells are significant anti-leukemic effector cells in patients with KIR/HLA genotypes that favor NK cell autoreactivity.
Collapse
Affiliation(s)
- E Bernson
- TIMM Laboratory, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - A Hallner
- TIMM Laboratory, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - F E Sander
- TIMM Laboratory, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - O Wilsson
- TIMM Laboratory, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - O Werlenius
- TIMM Laboratory, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden.,Department of Hematology, University of Gothenburg, Gothenburg, Sweden
| | - A Rydström
- TIMM Laboratory, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - R Kiffin
- TIMM Laboratory, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - M Brune
- Department of Hematology, University of Gothenburg, Gothenburg, Sweden
| | - R Foà
- Department of Cellular Biotechnologies and Hematology, Sapienza University, Rome, Italy
| | - J Aurelius
- TIMM Laboratory, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden.,Department of Hematology, University of Gothenburg, Gothenburg, Sweden
| | - A Martner
- TIMM Laboratory, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - K Hellstrand
- TIMM Laboratory, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - F B Thorén
- TIMM Laboratory, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
61
|
Cogdill AP, Andrews MC, Wargo JA. Hallmarks of response to immune checkpoint blockade. Br J Cancer 2017; 117:1-7. [PMID: 28524159 PMCID: PMC5520201 DOI: 10.1038/bjc.2017.136] [Citation(s) in RCA: 180] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 03/09/2017] [Accepted: 03/16/2017] [Indexed: 02/08/2023] Open
Abstract
Unprecedented advances have been made in the treatment of cancer through the use of immune checkpoint blockade, with approval of several checkpoint blockade regimens spanning multiple cancer types. However, responses to this form of therapy are not universal, and insights are clearly needed to identify optimal biomarkers of response and to combat mechanisms of therapeutic resistance. A working knowledge of the hallmarks of cancer yields insight into responses to immune checkpoint blockade, although the focus of this is rather tumour-centric and additional factors are pertinent, including host immunity and environmental influences. Herein, we describe the foundation for pillars and hallmarks of response to immune checkpoint blockade, with a discussion of their relevance to immune monitoring and mechanisms of resistance. Evolution of this understanding will ultimately help guide treatment strategies to enhance therapeutic responses.
Collapse
Affiliation(s)
- Alexandria P Cogdill
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Miles C Andrews
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Jennifer A Wargo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA.,Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| |
Collapse
|
62
|
Rathmann S, Keck C, Kreutz C, Weit N, Müller M, Timmer J, Glatzel S, Follo M, Malkovsky M, Werner M, Handgretinger R, Finke J, Fisch P. Partial break in tolerance of NKG2A−/LIR-1− single KIR+ NK cells early in the course of HLA-matched, KIR-mismatched hematopoietic cell transplantation. Bone Marrow Transplant 2017; 52:1144-1155. [DOI: 10.1038/bmt.2017.81] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 02/17/2017] [Accepted: 03/02/2017] [Indexed: 02/03/2023]
|
63
|
Baggio L, Laureano ÁM, Silla LMDR, Lee DA. Natural killer cell adoptive immunotherapy: Coming of age. Clin Immunol 2017; 177:3-11. [DOI: 10.1016/j.clim.2016.02.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 02/06/2016] [Accepted: 02/09/2016] [Indexed: 11/26/2022]
|
64
|
Whittle SB, Smith V, Doherty E, Zhao S, McCarty S, Zage PE. Overview and recent advances in the treatment of neuroblastoma. Expert Rev Anticancer Ther 2017; 17:369-386. [PMID: 28142287 DOI: 10.1080/14737140.2017.1285230] [Citation(s) in RCA: 258] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Children with neuroblastoma have widely divergent outcomes, ranging from cure in >90% of patients with low risk disease to <50% for those with high risk disease. Recent research has shed light on the biology of neuroblastoma, allowing for more accurate risk stratification and treatment reduction in many cases, although newer treatment strategies for children with high-risk and relapsed neuroblastoma are needed to improve outcomes. Areas covered: Neuroblastoma epidemiology, diagnosis, risk stratification, and recent advances in treatment of both newly diagnosed and relapsed neuroblastoma. Expert commentary: The identification of newer tumor targets and of novel cell-mediated immunotherapy agents may lead to novel therapeutic approaches, and clinical trials for regimens designed to target individual genetic aberrations in tumors are underway. A combination of therapeutic modalities will likely be required to improve survival and cure rates for patients with high-risk neuroblastoma.
Collapse
Affiliation(s)
- Sarah B Whittle
- a Department of Pediatrics, Section of Hematology-Oncology , Texas Children's Cancer and Hematology Centers, Baylor College of Medicine , Houston , TX , USA
| | - Valeria Smith
- a Department of Pediatrics, Section of Hematology-Oncology , Texas Children's Cancer and Hematology Centers, Baylor College of Medicine , Houston , TX , USA
| | - Erin Doherty
- a Department of Pediatrics, Section of Hematology-Oncology , Texas Children's Cancer and Hematology Centers, Baylor College of Medicine , Houston , TX , USA
| | - Sibo Zhao
- a Department of Pediatrics, Section of Hematology-Oncology , Texas Children's Cancer and Hematology Centers, Baylor College of Medicine , Houston , TX , USA
| | - Scott McCarty
- b Department of Pediatrics, Division of Hematology-Oncology , University of California San Diego, La Jolla, CA and Peckham Center for Cancer and Blood Disorders, Rady Children's Hospital , San Diego , CA , USA
| | - Peter E Zage
- b Department of Pediatrics, Division of Hematology-Oncology , University of California San Diego, La Jolla, CA and Peckham Center for Cancer and Blood Disorders, Rady Children's Hospital , San Diego , CA , USA
| |
Collapse
|
65
|
Leone P, De Re V, Vacca A, Dammacco F, Racanelli V. Cancer treatment and the KIR-HLA system: an overview. Clin Exp Med 2017; 17:419-429. [PMID: 28188495 DOI: 10.1007/s10238-017-0455-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 01/29/2017] [Indexed: 12/18/2022]
Abstract
Accumulating evidence indicates that the success of cancer therapy depends not only on a combination of adequate procedures (surgery, chemotherapy and radiotherapy) that aim to eliminate all tumor cells, but also on the functional state of the host immune system. HLA and KIR molecules, in particular, are critical to the interactions between tumor cells and both innate and adaptive immune cells such as NK cells and T cells. Different KIR-HLA gene combinations as well as different HLA expression levels on tumor cells associate with variable tumor prognosis and response to treatment. On the other hand, different therapies have different effects on HLA molecules and immune cell functions regulated by these molecules. Here, we provide an overview of the KIR-HLA system, a description of its alterations with clinical relevance in diverse tumor types, and an analysis of the consequences that conventional cancer therapies may have on it. We also discuss how this knowledge can be exploited to identify potential immunological biomarkers that can help to select patients for tailored therapy.
Collapse
Affiliation(s)
- Patrizia Leone
- Department of Internal Medicine and Clinical Oncology, University of Bari Medical School, Policlinico - 11, Piazza G. Cesare, 70124, Bari, Italy
| | - Valli De Re
- Bio-Proteomics Facility, Department of Translational Research, Centro di Riferimento Oncologico, National Cancer Institute, Aviano, Italy
| | - Angelo Vacca
- Department of Internal Medicine and Clinical Oncology, University of Bari Medical School, Policlinico - 11, Piazza G. Cesare, 70124, Bari, Italy
| | - Franco Dammacco
- Department of Internal Medicine and Clinical Oncology, University of Bari Medical School, Policlinico - 11, Piazza G. Cesare, 70124, Bari, Italy
| | - Vito Racanelli
- Department of Internal Medicine and Clinical Oncology, University of Bari Medical School, Policlinico - 11, Piazza G. Cesare, 70124, Bari, Italy.
| |
Collapse
|
66
|
Abstract
Neuroblastoma is the most common extracranial solid tumour occurring in childhood and has a diverse clinical presentation and course depending on the tumour biology. Unique features of these neuroendocrine tumours are the early age of onset, the high frequency of metastatic disease at diagnosis and the tendency for spontaneous regression of tumours in infancy. The most malignant tumours have amplification of the MYCN oncogene (encoding a transcription factor), which is usually associated with poor survival, even in localized disease. Although transgenic mouse models have shown that MYCN overexpression can be a tumour-initiating factor, many other cooperating genes and tumour suppressor genes are still under investigation and might also have a role in tumour development. Segmental chromosome alterations are frequent in neuroblastoma and are associated with worse outcome. The rare familial neuroblastomas are usually associated with germline mutations in ALK, which is mutated in 10-15% of primary tumours, and provides a potential therapeutic target. Risk-stratified therapy has facilitated the reduction of therapy for children with low-risk and intermediate-risk disease. Advances in therapy for patients with high-risk disease include intensive induction chemotherapy and myeloablative chemotherapy, followed by the treatment of minimal residual disease using differentiation therapy and immunotherapy; these have improved 5-year overall survival to 50%. Currently, new approaches targeting the noradrenaline transporter, genetic pathways and the tumour microenvironment hold promise for further improvements in survival and long-term quality of life.
Collapse
|
67
|
Kushner BH, Ostrovnaya I, Cheung IY, Kuk D, Modak S, Kramer K, Roberts SS, Basu EM, Yataghene K, Cheung NKV. Lack of survival advantage with autologous stem-cell transplantation in high-risk neuroblastoma consolidated by anti-GD2 immunotherapy and isotretinoin. Oncotarget 2016; 7:4155-66. [PMID: 26623730 PMCID: PMC4826196 DOI: 10.18632/oncotarget.6393] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 11/09/2015] [Indexed: 11/25/2022] Open
Abstract
Since 2003, high-risk neuroblastoma (HR-NB) patients at our center received anti-GD2 antibody 3F8/GM-CSF + isotretinoin – but not myeloablative therapy with autologous stem-cell transplantation (ASCT). Post-ASCT patients referred from elsewhere also received 3F8/GM-CSF + isotretinoin. We therefore accrued a study population of two groups treated during the same period and whose consolidative therapy, aside from ASCT, was identical. We analyzed patients enrolled in 1st complete/very good partial remission (CR/VGPR). Their event-free survival (EFS) and overall survival (OS) were calculated from study entry. Large study size allowed robust statistical analyses of key prognosticators including MYCN amplification, minimal residual disease (MRD), FCGR2A polymorphisms, and killer immunoglobulin-like receptor genotypes of natural killer cells. The 170 study patients included 60 enrolled following ASCT and 110 following conventional chemotherapy. The two cohorts had similar clinical and biological features. Five-year rates for ASCT and non-ASCT patients were, respectively: EFS 65% vs. 51% (p = .128), and OS 76% vs. 75% (p = .975). In multivariate analysis, ASCT was not prognostic and only MRD-negativity after two cycles of 3F8/GM-CSF correlated with significantly improved EFS and OS. Although a trend towards better EFS is seen with ASCT, OS is near identical. Cure rates may be similar, as close surveillance detects localized relapse and effective salvage treatments are applied. ASCT may not be needed to improve outcome when anti-GD2 immunotherapy is used for consolidation after dose-intensive conventional chemotherapy.
Collapse
Affiliation(s)
- Brian H Kushner
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Irina Ostrovnaya
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Irene Y Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Deborah Kuk
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Shakeel Modak
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Kim Kramer
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Stephen S Roberts
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ellen M Basu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Karima Yataghene
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
68
|
Liu F, Duan X, Wan Z, Zang H, You S, Yang R, Liu H, Li D, Li J, Zhang Y, Xin S. Lower number and decreased function of natural killer cells in hepatitis B virus related acute-on-chronic liver failure. Clin Res Hepatol Gastroenterol 2016; 40:605-613. [PMID: 27053076 DOI: 10.1016/j.clinre.2016.01.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 01/07/2016] [Accepted: 01/21/2016] [Indexed: 02/04/2023]
Abstract
BACKGROUND Hepatitis B virus (HBV)-related acute-on-chronic liver failure (ACLF) refers to acute deterioration occurring in patients with chronic hepatitis B infected liver diseases. An abnormality in NK cells mediated cellular immunity is believed to be a contributing factor. We aimed to evaluate the characteristic of NK cells in the peripheral blood of HBV related ACLF. METHODS Flow cytometric method was used to detect the absolute numbers and subgroups of NK cells, and analyze the cytotoxicity and killing ability of NK cells in patients with HBV-ACLF. RESULTS The results showed that peripheral numbers of NK cells were decreased in patients with HBV-ACLF, but not statistically significant. The cytotoxic CD56dimCD16bright NK cells were significantly decreased in HBV infected patients, especially ACLF patients. The CD56brightCD16- subgroup was expanded in patients with CHB and the CD56dimCD16- subgroup was expanded in patients with ACLF. The activating receptors of NKG2D, NKp30, NKp44, and NKp46 were increased in patients with ACLF. The inhibitory receptors of CD158a were increased, though the CD158b was decreased in patients of ACLF. The function of NK cells including cytotoxicity and killing activity were both downregulated in patients with ACLF and CHB. Even if after IL-12/15 stimulation, INF-γ and TNF-α produced by patients with ACLF were still less than those produced by healthy controls. CONCLUSIONS Patients with HBV-ACLF had lower numbers and decreased functions of cytotoxic NK cells.
Collapse
Affiliation(s)
| | | | | | - Hong Zang
- Beijing 302 Hospital, Beijing, China
| | | | | | | | - Dongze Li
- Beijing 302 Hospital, Beijing, China
| | - Jin Li
- Beijing 302 Hospital, Beijing, China
| | - Yawei Zhang
- Yale School of Public Health, Yale University, New Haven, CT, United States.
| | | |
Collapse
|
69
|
Yu AL, Hung JT, Ho MY, Yu J. Alterations of Glycosphingolipids in Embryonic Stem Cell Differentiation and Development of Glycan-Targeting Cancer Immunotherapy. Stem Cells Dev 2016; 25:1532-1548. [DOI: 10.1089/scd.2016.0138] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Alice L. Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Jung-Tung Hung
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan
| | - Ming-Yi Ho
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan
| | - John Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
70
|
Efficient Killing of High Risk Neuroblastoma Using Natural Killer Cells Activated by Plasmacytoid Dendritic Cells. PLoS One 2016; 11:e0164401. [PMID: 27716850 PMCID: PMC5055353 DOI: 10.1371/journal.pone.0164401] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 09/23/2016] [Indexed: 12/24/2022] Open
Abstract
High-risk neuroblastoma (NB) remains a major therapeutic challenge despite the recent advent of disialoganglioside (GD2)-antibody treatment combined with interleukin (IL)-2 and granulocyte monocyte-colony stimulating factor (GM-CSF). Indeed, more than one third of the patients still die from this disease. Here, we developed a novel approach to improve the current anti-GD2 immunotherapy based on NK cell stimulation using toll-like receptor (TLR)-activated plasmacytoid dendritic cells (pDCs). We demonstrated that this strategy led to the efficient killing of NB cells. When the expression of GD2 was heterogeneous on NB cells, the combination of pDC-mediated NK-cell activation and anti-GD2 treatment significantly increased the cytotoxicity of NK cells against NB cells. Activation by pDCs led to a unique NK-cell phenotype characterized by increased surface expression of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), with increased expression of CD69 on CD56dim cytotoxic cells, and strong interferon-γ production. Additionally, NB-cell killing was mediated by the TRAIL death-receptor pathway, as well as by the release of cytolytic granules via the DNAX accessory molecule 1 pathway. NK-cell activation and lytic activity against NB was independent of cell contact, depended upon type I IFN produced by TLR-9-activated pDCs, but was not reproduced by IFN-α stimulation alone. Collectively, these results highlighted the therapeutic potential of activated pDCs for patients with high-risk NB.
Collapse
|
71
|
Wang W, Erbe AK, Gallenberger M, Kim K, Carmichael L, Hess D, Mendonca EA, Song Y, Hank JA, Cheng SC, Signoretti S, Atkins M, Carlson A, Weiss JM, Mier J, Panka D, McDermott DF, Sondel PM. Killer immunoglobulin-like receptor (KIR) and KIR-ligand genotype do not correlate with clinical outcome of renal cell carcinoma patients receiving high-dose IL2. Cancer Immunol Immunother 2016; 65:1523-1532. [PMID: 27695964 DOI: 10.1007/s00262-016-1904-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 09/21/2016] [Indexed: 01/13/2023]
Abstract
NK cells play a role in many cancer immunotherapies. NK cell activity is tightly regulated by killer immunoglobulin-like receptor (KIR) and KIR-ligand interactions. Inhibitory KIR-ligands have been identified as HLA molecules, while activating KIR-ligands are largely unknown. Individuals that have not inherited the corresponding KIR-ligand for at least one inhibitory KIR gene are termed the "KIR-ligand missing" genotype, and they are thought to have a subset of NK cells that express inhibitory KIRs for which the corresponding KIR-ligand is missing on autologous tissue, and thus will not be inhibited through KIR-ligand recognition. In some settings where an anticancer immunotherapeutic effect is likely mediated by NK cells, individuals with a KIR-ligand missing genotype have shown improved clinical outcome compared to individuals with an "all KIR-ligands present" genotype. In addition, patients receiving hematopoietic stem cell transplants for leukemia may do better if their donor has more activating KIR genes (i.e., KIR haplotype-B). In a recent multi-institution clinical trial of patients with metastatic renal cell carcinoma receiving high-dose IL2 (HD-IL2), 25 % of patients showed a complete or partial tumor response to this therapy. We genotyped KIR and KIR-ligand genes for these patients (n = 107) and tested whether KIR/KIR-ligand genotypes correlated with patient clinical outcomes. In these analyses, we did not find any significant association of KIR/KIR-ligand genotype (either KIR-ligand missing or the presence of KIR haplotype-B) with patient outcome in response to the HD-IL2 therapy.
Collapse
Affiliation(s)
- Wei Wang
- Department of Human Oncology, University of Wisconsin, 4159 WIMR Bldg. UW-Madison Campus, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Amy K Erbe
- Department of Human Oncology, University of Wisconsin, 4159 WIMR Bldg. UW-Madison Campus, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Mikayla Gallenberger
- Department of Human Oncology, University of Wisconsin, 4159 WIMR Bldg. UW-Madison Campus, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - KyungMann Kim
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, WI, USA
| | - Lakeesha Carmichael
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, WI, USA
| | - Dustin Hess
- Department of Human Oncology, University of Wisconsin, 4159 WIMR Bldg. UW-Madison Campus, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Eneida A Mendonca
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, WI, USA.,Department of Pediatrics, University of Wisconsin, Madison, WI, USA
| | - Yiqiang Song
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, WI, USA
| | - Jacquelyn A Hank
- Department of Human Oncology, University of Wisconsin, 4159 WIMR Bldg. UW-Madison Campus, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Su-Chun Cheng
- Department of Biostatistics, Dana Farber Cancer Institute, Boston, MA, USA
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Michael Atkins
- The Cytokine Working Group, Rockville, MD, USA.,Department of Medicine, Georgetown University, Washington, DC, USA
| | - Alexander Carlson
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | - James Mier
- The Cytokine Working Group, Rockville, MD, USA.,Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - David Panka
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - David F McDermott
- The Cytokine Working Group, Rockville, MD, USA.,Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Paul M Sondel
- Department of Human Oncology, University of Wisconsin, 4159 WIMR Bldg. UW-Madison Campus, 1111 Highland Avenue, Madison, WI, 53705, USA. .,Department of Pediatrics, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
72
|
Siebert N, Jensen C, Troschke-Meurer S, Zumpe M, Jüttner M, Ehlert K, Kietz S, Müller I, Lode HN. Neuroblastoma patients with high-affinity FCGR2A, -3A and stimulatory KIR 2DS2 treated by long-term infusion of anti-GD 2 antibody ch14.18/CHO show higher ADCC levels and improved event-free survival. Oncoimmunology 2016; 5:e1235108. [PMID: 27999754 DOI: 10.1080/2162402x.2016.1235108] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 09/07/2016] [Accepted: 09/07/2016] [Indexed: 12/11/2022] Open
Abstract
Polymorphisms in Fc-gamma-receptor (FCGR) genes as well as killer cell immunoglobulin-like receptor (KIR) and KIR ligand (KIRL) repertoires may influence antitumor effects of monoclonal antibodies (mAb). Here, we systematically analyzed high- and low-affinity FCGR2A and -3A genotypes as well as stimulating and inhibitory KIR/KIRL combinations in 53 neuroblastoma (NB) patients treated by long-term infusion (LTI) of anti-GD2 IgG1 Ab ch14.18/CHO using validated real-time PCR methods. Patients with high-affinity FCGR2A and -3A genotypes showed a higher level of Ab-dependent cell-mediated cytotoxicity (ADCC) on day 8 after the start of ch14.18/CHO and superior event-free survival (EFS) compared to patients with low FCGR genotypes. Similar observations were made for patients with stimulatory KIR/KIRL haplotype B (combination of KIR genes including activating receptor genes) compared to inhibitory haplotype A (a fixed set of genes encoding for inhibitory receptors, except 2DS4) and stronger effects were found in patients when haplotype B and high-affinity FCGRs were combined. Surprisingly, independent analysis of KIRs showed a major role of activating KIR 2DS2 for high ADCC levels and prolongation of EFS. The greatest effect was observed in 2DS2-positive patients that also had high-affinity FCGR2A and -3A genotypes. In summary, the presence of the activating KIR 2DS2 has a major effect on ADCC levels and survival in NB patients treated by LTI of ch14.18/CHO and may therefore be a useful biomarker in combination with FCGR polymorphisms for Ab-based immunotherapies.
Collapse
Affiliation(s)
- Nikolai Siebert
- Department of Pediatric Oncology and Hematology, University Medicine Greifswald , Greifswald, Germany
| | - Christian Jensen
- Department of Pediatric Oncology and Hematology, University Medicine Greifswald , Greifswald, Germany
| | - Sascha Troschke-Meurer
- Department of Pediatric Oncology and Hematology, University Medicine Greifswald , Greifswald, Germany
| | - Maxi Zumpe
- Department of Pediatric Oncology and Hematology, University Medicine Greifswald , Greifswald, Germany
| | - Madlen Jüttner
- Department of Pediatric Oncology and Hematology, University Medicine Greifswald , Greifswald, Germany
| | - Karoline Ehlert
- Department of Pediatric Oncology and Hematology, University Medicine Greifswald , Greifswald, Germany
| | - Silke Kietz
- Department of Pediatric Oncology and Hematology, University Medicine Greifswald , Greifswald, Germany
| | - Ina Müller
- Department of Pediatric Oncology and Hematology, University Medicine Greifswald , Greifswald, Germany
| | - Holger N Lode
- Department of Pediatric Oncology and Hematology, University Medicine Greifswald , Greifswald, Germany
| |
Collapse
|
73
|
Wang W, Erbe AK, Alderson KA, Phillips E, Gallenberger M, Gan J, Campana D, Hank JA, Sondel PM. Human NK cells maintain licensing status and are subject to killer immunoglobulin-like receptor (KIR) and KIR-ligand inhibition following ex vivo expansion. Cancer Immunol Immunother 2016; 65:1047-59. [PMID: 27392940 PMCID: PMC5477646 DOI: 10.1007/s00262-016-1864-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 06/29/2016] [Indexed: 01/18/2023]
Abstract
Infusion of allogeneic NK cells is a potential immunotherapy for both hematopoietic malignancies and solid tumors. Interactions between killer immunoglobulin-like receptors (KIR) on human NK cells and KIR-ligands on tumor cells influence the magnitude of NK function. To obtain sufficient numbers of activated NK cells for infusion, one potent method uses cells from the K562 human erythroleukemia line that have been transfected to express activating 41BB ligand (41BBL) and membrane-bound interleukin 15 (mbIL15). The functional importance of KIRs on ex vivo expanded NK cells has not been studied in detail. We found that after a 12-day co-culture with K562-mbIL15-41BBL cells, expanded NK cells maintained inhibition specificity and prior in vivo licensing status determined by KIR/KIR-ligand interactions. Addition of an anti-CD20 antibody (rituximab) induced NK-mediated antibody-dependent cellular cytotoxicity and augmented killing of CD20+ target cells. However, partial inhibition induced by KIR/KIR-ligand interactions persisted. Finally, we found that extended co-cultures of NK cells with stimulatory cells transduced to express various KIR-ligands modified both the inhibitory and activating KIR repertoires of the expanded NK cell product. These studies demonstrate that the licensing interactions known to occur during NK ontogeny also influence NK cell function following NK expansion ex vivo with HLA-null stimulatory cells.
Collapse
Affiliation(s)
- Wei Wang
- Department of Human Oncology, University of Wisconsin, 4159 WIMR Bldg. UW-Madison Campus, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Amy K Erbe
- Department of Human Oncology, University of Wisconsin, 4159 WIMR Bldg. UW-Madison Campus, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Kory A Alderson
- Department of Human Oncology, University of Wisconsin, 4159 WIMR Bldg. UW-Madison Campus, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Emily Phillips
- Department of Human Oncology, University of Wisconsin, 4159 WIMR Bldg. UW-Madison Campus, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Mikayla Gallenberger
- Department of Human Oncology, University of Wisconsin, 4159 WIMR Bldg. UW-Madison Campus, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Jacek Gan
- Department of Human Oncology, University of Wisconsin, 4159 WIMR Bldg. UW-Madison Campus, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Dario Campana
- Department of Pediatrics, National University of Singapore, Singapore, Singapore
| | - Jacquelyn A Hank
- Department of Human Oncology, University of Wisconsin, 4159 WIMR Bldg. UW-Madison Campus, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Paul M Sondel
- Department of Human Oncology, University of Wisconsin, 4159 WIMR Bldg. UW-Madison Campus, 1111 Highland Avenue, Madison, WI, 53705, USA.
- Department of Pediatrics, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
74
|
Perez Horta Z, Goldberg JL, Sondel PM. Anti-GD2 mAbs and next-generation mAb-based agents for cancer therapy. Immunotherapy 2016; 8:1097-117. [PMID: 27485082 PMCID: PMC5619016 DOI: 10.2217/imt-2016-0021] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 05/11/2016] [Indexed: 12/16/2022] Open
Abstract
Tumor-specific monoclonal antibodies (mAbs) have demonstrated efficacy in the clinic, becoming an important approach for cancer immunotherapy. Due to its limited expression on normal tissue, the GD2 disialogangloside expressed on neuroblastoma cells is an excellent candidate for mAb therapy. In 2015, dinutuximab (an anti-GD2 mAb) was approved by the US FDA and is currently used in a combination immunotherapeutic regimen for the treatment of children with high-risk neuroblastoma. Here, we review the extensive preclinical and clinical development of anti-GD2 mAbs and the different mechanisms by which they mediate tumor cell killing. In addition, we discuss different mAb-based strategies that capitalize on the targeting ability of anti-GD2 mAbs to potentially deliver, as monotherapy, or in combination with other treatments, improved antitumor efficacy.
Collapse
Affiliation(s)
| | - Jacob L Goldberg
- Department of Human Oncology, University of Wisconsin, Madison, WI, USA
| | - Paul M Sondel
- Department of Human Oncology, University of Wisconsin, Madison, WI, USA
- Department of Pediatrics & Genetics, University of Wisconsin School of Medicine & Public Health, Madison, WI, USA
| |
Collapse
|
75
|
Barrett DM, Bagatell R. The Beginning of the End of Package Deal Therapy for Patients With High-Risk Neuroblastoma? J Clin Oncol 2016; 34:2437-9. [DOI: 10.1200/jco.2016.67.2774] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
|
76
|
Zhen Z, Guo X, Liao R, Yang K, Ye L, You Z. Involvement of IL-10 and TGF-β in HLA-E-mediated neuroblastoma migration and invasion. Oncotarget 2016; 7:44340-44349. [PMID: 27322426 PMCID: PMC5190101 DOI: 10.18632/oncotarget.10041] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 06/03/2016] [Indexed: 12/23/2022] Open
Abstract
Human leukocyte antigen (HLA)-E is highly expressed in a variety of tumors and, in addition to immune escape, may promote tumor growth via other mechanisms. However, the role of HLA-E in neuroblastoma (NB) migration and invasion is unknown. In the present study, HLA-E expression in human NB tumors was measured by immunohistochemistry. The effect of HLA-E on NB cell migration and invasion was studied in vitro and in vivo, as well as the effect of HLA-E on natural killer (NK)-cell cytotoxicity. HLA-E was expressed in 70.2% of the NB tumor tissues examined. HLA-E expression by NB cells inhibited NK-cell cytotoxicity and induced the release of interleukin (IL)-10 and transforming growth factor (TGF)-β1. HLA-E and the released cytokines enhanced the ability of NB cells migration and invasion. NK cell infusion did not inhibit the growth of NB cells with high HLA-E expression but instead increased the number of metastatic cells in the bone marrow. Taken together, the results indicate that IL-10 and TGF-β are involved in HLA-E-mediated NB migration and invasion. Thus, HLA-E may be a new treatment target in NB.
Collapse
Affiliation(s)
- Zijun Zhen
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- State Key Laboratory of Oncology in South China, Guangzhou, 510060, China
- Collaborative Innovation Center of Cancer Medicine, Guangzhou, 510060, China
| | - Xiaofang Guo
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- State Key Laboratory of Oncology in South China, Guangzhou, 510060, China
- Collaborative Innovation Center of Cancer Medicine, Guangzhou, 510060, China
| | - Ru Liao
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- State Key Laboratory of Oncology in South China, Guangzhou, 510060, China
- Collaborative Innovation Center of Cancer Medicine, Guangzhou, 510060, China
| | - Kaibin Yang
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Litong Ye
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Zhiyao You
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| |
Collapse
|
77
|
Challenges in the harmonization of immune monitoring studies and trial design for cell-based therapies in the context of hematopoietic cell transplantation for pediatric cancer patients. Cytotherapy 2016; 17:1667-74. [PMID: 26589751 DOI: 10.1016/j.jcyt.2015.09.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 09/15/2015] [Accepted: 09/21/2015] [Indexed: 12/16/2022]
Abstract
Clinical trials aimed at improving results of hematopoietic cell transplantation (HCT) by adjuvant cell-based interventions in children have been limited by small numbers and pediatric-specific features. The need for a larger number of pediatric HCT centers to participate in trials has resulted in a demand for harmonization of disease-specific clinical trials and immune-monitoring. Thus far, most phase I/II trials select different end points evaluated at disparate time points, making inter-study comparisons difficult and, sometimes, impossible. In this review, we discuss the various aspects that are important to consider for harmonizing clinical trial design as well as the critical elements for standardized (immune)-monitoring protocols in cell-based intervention trials in the context of HCT. Comparison data from trials applying harmonized trial design will lead to optimized immunotherapeutic treatment protocols to maximize clinical efficacy while minimizing toxicity.
Collapse
|
78
|
Xu H, Guo H, Cheung IY, Cheung NKV. Antitumor Efficacy of Anti-GD2 IgG1 Is Enhanced by Fc Glyco-Engineering. Cancer Immunol Res 2016; 4:631-8. [PMID: 27197064 DOI: 10.1158/2326-6066.cir-15-0221] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 03/29/2016] [Indexed: 12/14/2022]
Abstract
The affinity of therapeutic antibodies for Fcγ receptors (FcγRs) strongly influences their antitumor potency. To generate antibodies with optimal binding and immunologic efficacy, we compared the affinities of different versions of an IgG1 Fc region that had an altered peptide backbone, altered glycans, or both. To produce IgG1 with glycans that lacked α1,6-fucose, we used CHO cells that were deficient in the enzyme UDP-N-acetylglucosamine: α-3-d-mannoside-β-1,2-N-acetylglucosaminyltransferase I (GnT1), encoded by the MGAT1 gene. Mature N-linked glycans require this enzyme, and without it, CHO cells synthesize antibodies carrying only Man5-GlcNAc2, which were more effective in antibody-dependent cell-mediated cytotoxicity (ADCC). Our engineered IgG1, hu3F8-IgG1, is specific for GD2, a neuroendocrine tumor ganglioside. Its peptide mutant is IgG1-DEL (S239D/I332E/A330L), both produced in wild-type CHO cells. When produced in GnT1-deficient CHO cells, we refer to them as IgG1n and IgG1n-DEL, respectively. Affinities for human FcγRs were measured using Biacore T-100 (on CD16 and CD32 polymorphic alleles), their immunologic properties compared for ADCC and complement-mediated cytotoxicity (CMC) in vitro, and pharmacokinetics and antitumor effects were compared in vivo in humanized mice. IgG1n and IgG1n-DEL contained only mannose and acetylglucosamine and had preferential affinity for activating CD16s, over inhibitory CD32B, receptors. In vivo, the antitumor effects of IgG1, IgG1-DEL, and IgG1n-DEL were similar but modest, whereas IgG1n was significantly more effective (P < 0.05). Thus, IgG1n antibodies produced in GnT1-deficient CHO cells may have potential as improved anticancer therapeutics. Cancer Immunol Res; 4(7); 631-8. ©2016 AACR.
Collapse
Affiliation(s)
- Hong Xu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Hongfen Guo
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Irene Y Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
79
|
Morris ZS, Guy EI, Francis DM, Gressett MM, Werner LR, Carmichael LL, Yang RK, Armstrong EA, Huang S, Navid F, Gillies SD, Korman A, Hank JA, Rakhmilevich AL, Harari PM, Sondel PM. In Situ Tumor Vaccination by Combining Local Radiation and Tumor-Specific Antibody or Immunocytokine Treatments. Cancer Res 2016; 76:3929-41. [PMID: 27197149 DOI: 10.1158/0008-5472.can-15-2644] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 03/04/2016] [Indexed: 01/06/2023]
Abstract
Interest in combining radiotherapy and immune checkpoint therapy is growing rapidly. In this study, we explored a novel combination of this type to augment antitumor immune responses in preclinical murine models of melanoma, neuroblastoma, and head and neck squamous cell carcinoma. Cooperative effects were observed with local radiotherapy and intratumoral injection of tumor-specific antibodies, arising in part from enhanced antibody-dependent cell-mediated cytotoxicity (ADCC). We could improve this response by combining radiation with intratumoral injection of an IL2-linked tumor-specific antibody (termed here an immunocytokine), resulting in complete regression of established tumors in most animals associated with a tumor-specific memory T-cell response. Given the T-cell response elicited by combined local radiation and intratumoral immunocytokine, we tested the potential benefit of adding this treatment to immune checkpoint blockade. In mice bearing large primary tumors or disseminated metastases, the triple-combination of intratumoral immunocytokine, radiation, and systemic anti-CTLA-4 improved primary tumor response and animal survival compared with combinations of any two of these three interventions. Taken together, our results show how combining radiation and intratumoral immunocytokine in murine tumor models can eradicate large tumors and metastases, eliciting an in situ vaccination effect that can be leveraged further by T-cell checkpoint blockade, with immediate implications for clinical evaluation. Cancer Res; 76(13); 3929-41. ©2016 AACR.
Collapse
Affiliation(s)
- Zachary S Morris
- Department of Human Oncology, University of Wisconsin, Madison, Wisconsin.
| | - Emily I Guy
- Department of Human Oncology, University of Wisconsin, Madison, Wisconsin
| | - David M Francis
- Department of Human Oncology, University of Wisconsin, Madison, Wisconsin
| | - Monica M Gressett
- Department of Human Oncology, University of Wisconsin, Madison, Wisconsin
| | - Lauryn R Werner
- Department of Human Oncology, University of Wisconsin, Madison, Wisconsin
| | - Lakeesha L Carmichael
- Department of Biostatistics and Bioinformatics, University of Wisconsin, Madison, Wisconsin
| | - Richard K Yang
- Department of Human Oncology, University of Wisconsin, Madison, Wisconsin
| | - Eric A Armstrong
- Department of Human Oncology, University of Wisconsin, Madison, Wisconsin
| | - Shyhmin Huang
- Department of Human Oncology, University of Wisconsin, Madison, Wisconsin
| | - Fariba Navid
- Department of Oncology, St. Jude Children's Hospital, Memphis, Tennessee
| | | | | | - Jacquelyn A Hank
- Department of Human Oncology, University of Wisconsin, Madison, Wisconsin
| | | | - Paul M Harari
- Department of Human Oncology, University of Wisconsin, Madison, Wisconsin
| | - Paul M Sondel
- Department of Human Oncology, University of Wisconsin, Madison, Wisconsin. Department of Pediatrics, University of Wisconsin, Madison, Wisconsin. Department of Genetics, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
80
|
Forlenza CJ, Boudreau JE, Zheng J, Le Luduec JB, Chamberlain E, Heller G, Cheung NKV, Hsu KC. KIR3DL1 Allelic Polymorphism and HLA-B Epitopes Modulate Response to Anti-GD2 Monoclonal Antibody in Patients With Neuroblastoma. J Clin Oncol 2016; 34:2443-51. [PMID: 27069083 DOI: 10.1200/jco.2015.64.9558] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
PURPOSE In patients with neuroblastoma (NB), treatment with anti-GD2 monoclonal antibody (mAb) directs natural killer (NK) cell-mediated antibody-dependent cellular cytotoxicity (ADCC) against tumor cells. However, tumor cytotoxicity is attenuated by ligation of inhibitory killer immunoglobulin-like receptors (KIRs) by HLA class I molecules. KIR3DL1 polymorphism influences its ability to engage HLA-Bw4 ligands. We tested the hypothesis that poorly interacting combinations of KIR3DL1 and HLA ligands are more permissive of mAb-mediated antitumor effect. METHODS KIR3DL1 and HLA-B subtyping were performed with a multiplex intermediate-resolution polymerase chain reaction assay for a cohort of 245 patients who were treated with antibody 3F8 for high-risk NB. Patient outcomes were analyzed according to expected degree of interaction between KIR3DL1 and HLA-B subtypes and grouped as strong, weak, or noninteractors. A comparison of NK response to 3F8 mAb opsonized NB cells between strong- and noninteracting donors was performed by flow cytometry. RESULTS KIR3DL1 and HLA-B subtype combinations associated with noninteraction as a result of lack of receptor expression [KIR3DL1(-)], failure of interaction with inhibitory ligands [KIR3DS1(+)], or absence of KIR ligands resulted in significantly improved overall and progression-free survival. Patients with KIR3DL1 and HLA-B subtype combinations that were predictive of weak interaction had superior outcomes compared with those that were predictive of strong interaction; however, both groups were inferior to those with noninteracting subtype combinations. In vitro analysis of 3F8-mediated ADCC showed that KIR3DL1(-) and 3DS1(+) NK cells were insensitive to inhibition by HLA-Bw4-expressing NB targets. CONCLUSION We conclude that KIR3LD1 and HLA-B allele combinations can have a prognostic impact on patient survival after treatment with anti-GD2 mAb that relies on NK-ADCC. The survival advantage seen in noninteracting combinations supports the therapeutic disinhibition of individuals with strongly interacting KIR and ligand pairs.
Collapse
Affiliation(s)
- Christopher J Forlenza
- Christopher J. Forlenza, Jeanette E. Boudreau, Junting Zheng, Jean-Benoît Le Luduec, Elizabeth Chamberlain, Glenn Heller, Nai-Kong V. Cheung, and Katharine C. Hsu, Memorial Sloan Kettering Cancer Center; and Katharine C. Hsu, Weill Cornell Medical College, New York, NY
| | - Jeanette E Boudreau
- Christopher J. Forlenza, Jeanette E. Boudreau, Junting Zheng, Jean-Benoît Le Luduec, Elizabeth Chamberlain, Glenn Heller, Nai-Kong V. Cheung, and Katharine C. Hsu, Memorial Sloan Kettering Cancer Center; and Katharine C. Hsu, Weill Cornell Medical College, New York, NY
| | - Junting Zheng
- Christopher J. Forlenza, Jeanette E. Boudreau, Junting Zheng, Jean-Benoît Le Luduec, Elizabeth Chamberlain, Glenn Heller, Nai-Kong V. Cheung, and Katharine C. Hsu, Memorial Sloan Kettering Cancer Center; and Katharine C. Hsu, Weill Cornell Medical College, New York, NY
| | - Jean-Benoît Le Luduec
- Christopher J. Forlenza, Jeanette E. Boudreau, Junting Zheng, Jean-Benoît Le Luduec, Elizabeth Chamberlain, Glenn Heller, Nai-Kong V. Cheung, and Katharine C. Hsu, Memorial Sloan Kettering Cancer Center; and Katharine C. Hsu, Weill Cornell Medical College, New York, NY
| | - Elizabeth Chamberlain
- Christopher J. Forlenza, Jeanette E. Boudreau, Junting Zheng, Jean-Benoît Le Luduec, Elizabeth Chamberlain, Glenn Heller, Nai-Kong V. Cheung, and Katharine C. Hsu, Memorial Sloan Kettering Cancer Center; and Katharine C. Hsu, Weill Cornell Medical College, New York, NY
| | - Glenn Heller
- Christopher J. Forlenza, Jeanette E. Boudreau, Junting Zheng, Jean-Benoît Le Luduec, Elizabeth Chamberlain, Glenn Heller, Nai-Kong V. Cheung, and Katharine C. Hsu, Memorial Sloan Kettering Cancer Center; and Katharine C. Hsu, Weill Cornell Medical College, New York, NY
| | - Nai-Kong V Cheung
- Christopher J. Forlenza, Jeanette E. Boudreau, Junting Zheng, Jean-Benoît Le Luduec, Elizabeth Chamberlain, Glenn Heller, Nai-Kong V. Cheung, and Katharine C. Hsu, Memorial Sloan Kettering Cancer Center; and Katharine C. Hsu, Weill Cornell Medical College, New York, NY
| | - Katharine C Hsu
- Christopher J. Forlenza, Jeanette E. Boudreau, Junting Zheng, Jean-Benoît Le Luduec, Elizabeth Chamberlain, Glenn Heller, Nai-Kong V. Cheung, and Katharine C. Hsu, Memorial Sloan Kettering Cancer Center; and Katharine C. Hsu, Weill Cornell Medical College, New York, NY.
| |
Collapse
|
81
|
Neri D, Sondel PM. Immunocytokines for cancer treatment: past, present and future. Curr Opin Immunol 2016; 40:96-102. [PMID: 27060634 DOI: 10.1016/j.coi.2016.03.006] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 03/15/2016] [Accepted: 03/15/2016] [Indexed: 12/11/2022]
Abstract
Immunocytokines are antibody-cytokine fusion proteins, with the potential to preferentially localize on tumor lesions and to activate anticancer immunity at the site of disease. Various tumor targets (e.g., cell membrane antigens and extracellular matrix components) and antibody formats (e.g., intact IgG and antibody fragments) have been considered for immunocytokine development and some products have advanced to clinical trials. In this review, we present relevant concepts and strategies for the design and use of anticancer immunocytokine products. In addition, we discuss emerging strategies for the pharmaceutical development and clinical application of this promising class of biopharmaceuticals.
Collapse
Affiliation(s)
- Dario Neri
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Vladimir-Prelog-Weg 4, CH-8093 Zürich, Switzerland.
| | - Paul M Sondel
- Departments of Pediatrics, Human Oncology and Genetics, and UW Carbone Cancer Center, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
82
|
Mise N, Takami M, Suzuki A, Kamata T, Harada K, Hishiki T, Saito T, Terui K, Mitsunaga T, Nakata M, Ikeuchi T, Nakayama T, Yoshida H, Motohashi S. Antibody-dependent cellular cytotoxicity toward neuroblastoma enhanced by activated invariant natural killer T cells. Cancer Sci 2016; 107:233-41. [PMID: 26749374 PMCID: PMC4814252 DOI: 10.1111/cas.12882] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 12/15/2015] [Accepted: 01/03/2016] [Indexed: 01/10/2023] Open
Abstract
Anti‐ganglioside GD2 antibodies mainly work through antibody‐dependent cellular cytotoxicity (ADCC) and have demonstrated clinical benefit for children with neuroblastoma. However, high‐risk neuroblastoma still has a high recurrence rate. For further improvement in patient outcomes, ways to maximize the cytotoxic effects of anti‐GD2 therapies with minimal toxicity are required. Activated invariant natural killer T (iNKT) cells enhance both innate and type I acquired anti‐tumor immunity by producing several kinds of cytokines. In this report, we investigated the feasibility of combination therapy using iNKT cells and an anti‐GD2 antibody. Although some of the expanded iNKT cells expressed natural killer (NK) cell markers, including FcγR, iNKT cells were not directly associated with ADCC. When co‐cultured with activated iNKT cells, granzyme A, granzyme B and interferon gamma (IFNγ) production from NK cells were upregulated, and the cytotoxicity of NK cells treated with anti‐GD2 antibodies was increased. Not only cytokines produced by activated iNKT cells, but also NK‐NKT cell contact or NK cell‐dendritic cell contact contributed to the increase in NK cell cytotoxicity and further IFNγ production by iNKT cells and NK cells. In conclusion, iNKT cell‐based immunotherapy could be an appropriate candidate for anti‐GD2 antibody therapy for neuroblastoma.
Collapse
Affiliation(s)
- Naoko Mise
- Department of Medical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan.,Department of Pediatric Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Mariko Takami
- Department of Medical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Akane Suzuki
- Department of Medical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Toshiko Kamata
- Department of Medical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kazuaki Harada
- Department of Medical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan.,Department of Pediatric Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Tomoro Hishiki
- Department of Pediatric Surgery, Chiba Children's Hospital, Chiba, Japan
| | - Takeshi Saito
- Department of Pediatric Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Keita Terui
- Department of Pediatric Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Tetsuya Mitsunaga
- Department of Pediatric Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Mitsuyuki Nakata
- Department of Pediatric Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Takayuki Ikeuchi
- Center for Advanced Medicine, Chiba University Hospital, Chiba, Japan
| | - Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hideo Yoshida
- Department of Pediatric Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Shinichiro Motohashi
- Department of Medical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
83
|
Abstract
PURPOSE OF REVIEW Major advances in our understanding of the genetic basis of neuroblastoma, and the role somatic alterations play in driving tumor growth, have led to improvements in risk-stratified therapy and have provided the rationale for targeted therapies. In this review, we highlight current risk-based treatment approaches and discuss the opportunities and challenges of translating recent genomic discoveries into the clinic. RECENT FINDINGS Significant progress in the treatment of neuroblastoma has been realized using risk-based treatment strategies. Outcome has improved for all patients, including those classified as high-risk, although survival remains poor for this cohort. Integration of whole-genome DNA copy number and comprehensive molecular profiles into neuroblastoma classification systems will allow more precise prognostication and refined treatment assignment. Promising treatments that include targeted systemic radiotherapy, pathway-targeted small molecules, and therapy targeted at cell surface molecules are being evaluated in clinical trials, and recent genomic discoveries in relapsed tumor samples have led to the identification of new actionable mutations. SUMMARY The integration of refined treatment stratification based on whole-genome profiles with therapeutics that target the molecular drivers of malignant behavior in neuroblastoma has the potential to dramatically improve survival, with decreased toxicity.
Collapse
|
84
|
Croce M, Corrias MV, Rigo V, Ferrini S. New immunotherapeutic strategies for the treatment of neuroblastoma. Immunotherapy 2016; 7:285-300. [PMID: 25804480 DOI: 10.2217/imt.14.117] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The prognosis of high-risk neuroblastoma (NB) is still poor, in spite of aggressive multimodal treatment. Recently, adjuvant immunotherapy with anti-GD2 antibodies combined with IL-2 or GM-CSF has been shown to improve survival. Several other immunotherapy strategies proved efficacy in preclinical models of NB, including different types of vaccines, adoptive cell therapies and combined approaches. The remarkable differences in the immunobiology of syngeneic models and human NB may, at least in part, limit the translation of preclinical therapies to a clinical setting. Nonetheless, several preliminary evidences suggest that new antibodies, cancer vaccines and adoptive transfer of lymphocytes, genetically engineered to acquire NB specificity, may result in clinical benefit, and clinical studies are currently ongoing.
Collapse
Affiliation(s)
- Michela Croce
- IRCCS-A.O.U. San-Martino-IST Istituto Nazionale per la Ricerca sul Cancro, Biotherapy Unit c/o CBA Torre C2, Largo R. Benzi 10, 16132 Genoa, Italy
| | | | | | | |
Collapse
|
85
|
Semeraro M, Rusakiewicz S, Minard-Colin V, Delahaye NF, Enot D, Vély F, Marabelle A, Papoular B, Piperoglou C, Ponzoni M, Perri P, Tchirkov A, Matta J, Lapierre V, Shekarian T, Valsesia-Wittmann S, Commo F, Prada N, Poirier-Colame V, Bressac B, Cotteret S, Brugieres L, Farace F, Chaput N, Kroemer G, Valteau-Couanet D, Zitvogel L. Clinical impact of the NKp30/B7-H6 axis in high-risk neuroblastoma patients. Sci Transl Med 2016; 7:283ra55. [PMID: 25877893 DOI: 10.1126/scitranslmed.aaa2327] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The immunosurveillance mechanisms governing high-risk neuroblastoma (HR-NB), a major pediatric malignancy, have been elusive. We identify a potential role for natural killer (NK) cells, in particular the interaction between the NK receptor NKp30 and its ligand, B7-H6, in the metastatic progression and survival of HR-NB after myeloablative multimodal chemotherapy and stem cell transplantation. NB cells expressing the NKp30 ligand B7-H6 stimulated NK cells in an NKp30-dependent manner. Serum concentration of soluble B7-H6 correlated with the down-regulation of NKp30, bone marrow metastases, and chemoresistance, and soluble B7-H6 contained in the serum of HR-NB patients inhibited NK cell functions in vitro. The expression of distinct NKp30 isoforms affecting the polarization of NK cell functions correlated with 10-year event-free survival in three independent cohorts of HR-NB in remission from metastases after induction chemotherapy (n = 196, P < 0.001), adding prognostic value to known risk factors such as N-Myc amplification and age >18 months. We conclude that the interaction between NKp30 and B7-H6 may contribute to the fate of NB patients and that both the expression of NKp30 isoforms on circulating NK cells and the concentration of soluble B7-H6 in the serum may be clinically useful as biomarkers for risk stratification.
Collapse
Affiliation(s)
- Michaela Semeraro
- Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), 114 rue Edouard Vaillant, 94805 Villejuif, France. INSERM U1015, GRCC, 94805 Villejuif, France. Department of Pediatric Oncology, GRCC, 94805 Villejuif, France. University of Paris Sud XI, 94805 Villejuif, France. Equipe 11 labelisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, 75006 Paris, France. INSERM U1138, 94805 Villejuif, France
| | - Sylvie Rusakiewicz
- Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), 114 rue Edouard Vaillant, 94805 Villejuif, France. INSERM U1015, GRCC, 94805 Villejuif, France. Center of Clinical Investigations in Biotherapies of Cancer, CICBT507, GRCC, 94805 Villejuif, France
| | - Véronique Minard-Colin
- Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), 114 rue Edouard Vaillant, 94805 Villejuif, France. INSERM U1015, GRCC, 94805 Villejuif, France. Department of Pediatric Oncology, GRCC, 94805 Villejuif, France
| | - Nicolas F Delahaye
- Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), 114 rue Edouard Vaillant, 94805 Villejuif, France. INSERM U1015, GRCC, 94805 Villejuif, France
| | - David Enot
- Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), 114 rue Edouard Vaillant, 94805 Villejuif, France. Equipe 11 labelisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, 75006 Paris, France. INSERM U1138, 94805 Villejuif, France
| | - Frédéric Vély
- Centre d'Immunologie de Marseille-Luminy, INSERM, U1104, F-13009 Marseille, France. CNRS, UMR7280, F-13009 Marseille, France. Aix Marseille Université, UM2, F-13009 Marseille, France. Service d'Immunologie, Assistance Publique-Hôpitaux de Marseille, Hôpital de la Conception, F-13009 Marseille, France
| | - Aurélien Marabelle
- Centre de Recherche en Cancérologie de Lyon, UMR INSERM U1052 CNRS 5286, Centre Léon Bérard, Université de Lyon, 69000 Lyon, France
| | - Benjamin Papoular
- Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), 114 rue Edouard Vaillant, 94805 Villejuif, France. INSERM U1015, GRCC, 94805 Villejuif, France
| | - Christelle Piperoglou
- Service d'Immunologie, Assistance Publique-Hôpitaux de Marseille, Hôpital de la Conception, F-13009 Marseille, France
| | - Mirco Ponzoni
- Giannina Gaslini Hospital, Experimental Therapy Unit Laboratory of Oncology, 16147 Genoa, Italy
| | - Patrizia Perri
- Giannina Gaslini Hospital, Experimental Therapy Unit Laboratory of Oncology, 16147 Genoa, Italy
| | - Andrei Tchirkov
- EA 4677 ERTICa, CHU et Centre Jean Perrin, 63011 Clermont-Ferrand, France. CHU de Clermont-Ferrand, Service de Cytogénétique Médicale, Hôpital Estaing, 63001 Clermont-Ferrand, France
| | - Jessica Matta
- Centre d'Immunologie de Marseille-Luminy, INSERM, U1104, F-13009 Marseille, France. CNRS, UMR7280, F-13009 Marseille, France. Aix Marseille Université, UM2, F-13009 Marseille, France
| | - Valérie Lapierre
- Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), 114 rue Edouard Vaillant, 94805 Villejuif, France. Cell Therapy Unit, GRCC, 94805 Villejuif, France
| | - Tala Shekarian
- Centre de Recherche en Cancérologie de Lyon, UMR INSERM U1052 CNRS 5286, Centre Léon Bérard, Université de Lyon, 69000 Lyon, France
| | - Sandrine Valsesia-Wittmann
- Centre de Recherche en Cancérologie de Lyon, UMR INSERM U1052 CNRS 5286, Centre Léon Bérard, Université de Lyon, 69000 Lyon, France
| | - Frédéric Commo
- Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), 114 rue Edouard Vaillant, 94805 Villejuif, France. INSERM U1015, GRCC, 94805 Villejuif, France
| | - Nicole Prada
- Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), 114 rue Edouard Vaillant, 94805 Villejuif, France. INSERM U1015, GRCC, 94805 Villejuif, France
| | - Vichnou Poirier-Colame
- Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), 114 rue Edouard Vaillant, 94805 Villejuif, France. INSERM U1015, GRCC, 94805 Villejuif, France
| | - Brigitte Bressac
- Service de Génétique, Molecular Genetic Department, GRCC, 94805 Villejuif, France
| | - Sophie Cotteret
- Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), 114 rue Edouard Vaillant, 94805 Villejuif, France. INSERM U1015, GRCC, 94805 Villejuif, France
| | - Laurence Brugieres
- Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), 114 rue Edouard Vaillant, 94805 Villejuif, France. Department of Pediatric Oncology, GRCC, 94805 Villejuif, France
| | - Françoise Farace
- Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), 114 rue Edouard Vaillant, 94805 Villejuif, France. INSERM U981, 94805 Villejuif, France
| | - Nathalie Chaput
- Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), 114 rue Edouard Vaillant, 94805 Villejuif, France. INSERM U1015, GRCC, 94805 Villejuif, France. Center of Clinical Investigations in Biotherapies of Cancer, CICBT507, GRCC, 94805 Villejuif, France
| | - Guido Kroemer
- Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), 114 rue Edouard Vaillant, 94805 Villejuif, France. Equipe 11 labelisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, 75006 Paris, France. INSERM U1138, 94805 Villejuif, France. University of Paris Descartes/ParisV, Sorbonne Paris Cité, 75005 Paris, France. Pôle de Biologie, Hôpital Européen Georges Pompidou, 75015 Paris, France.
| | - Dominique Valteau-Couanet
- Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), 114 rue Edouard Vaillant, 94805 Villejuif, France. INSERM U1015, GRCC, 94805 Villejuif, France. Department of Pediatric Oncology, GRCC, 94805 Villejuif, France
| | - Laurence Zitvogel
- Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), 114 rue Edouard Vaillant, 94805 Villejuif, France. INSERM U1015, GRCC, 94805 Villejuif, France. University of Paris Sud XI, 94805 Villejuif, France. Center of Clinical Investigations in Biotherapies of Cancer, CICBT507, GRCC, 94805 Villejuif, France.
| |
Collapse
|
86
|
Abstract
Natural killer (NK) cells play a central role in immune responses through direct cytotoxicity and the release of cytokines that prime adaptive immunity. In simian primates, NK cell responses are regulated by interactions between two highly polymorphic sets of molecules: the killer-cell immunoglobulin-like receptors (KIRs) and their major histocompatibility complex (MHC) class I ligands. KIR-MHC class I interactions in humans have been implicated in the outcome of a number viral diseases and cancers. However, studies to address the role of KIRs in animal models have been limited by the complex immunogenetics and lack of defined ligands for KIRs in non-human primates. Due to the rapid evolution of KIRs, there is little conservation among the KIR genes of different primate species and it is not possible to predict the specificity of KIRs from known KIR-MHC class I interactions in humans. Hence, the MHC class I ligands for KIRs in species other than humans are poorly defined. Here, we review the KIR genes of the rhesus macaque, an important animal model for human immunodeficiency virus infection and other infectious diseases, and the MHC class I ligands that have been identified for KIRs in this species.
Collapse
Affiliation(s)
- Benjamin N. Bimber
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR 97239
| | - David T. Evans
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53711
| |
Collapse
|
87
|
Wang W, Erbe AK, Hank JA, Morris ZS, Sondel PM. NK Cell-Mediated Antibody-Dependent Cellular Cytotoxicity in Cancer Immunotherapy. Front Immunol 2015; 6:368. [PMID: 26284063 PMCID: PMC4515552 DOI: 10.3389/fimmu.2015.00368] [Citation(s) in RCA: 384] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 07/06/2015] [Indexed: 12/21/2022] Open
Abstract
Natural killer (NK) cells play a major role in cancer immunotherapies that involve tumor-antigen targeting by monoclonal antibodies (mAbs). NK cells express a variety of activating and inhibitory receptors that serve to regulate the function and activity of the cells. In the context of targeting cells, NK cells can be "specifically activated" through certain Fc receptors that are expressed on their cell surface. NK cells can express FcγRIIIA and/or FcγRIIC, which can bind to the Fc portion of immunoglobulins, transmitting activating signals within NK cells. Once activated through Fc receptors by antibodies bound to target cells, NK cells are able to lyse target cells without priming, and secrete cytokines like interferon gamma to recruit adaptive immune cells. This antibody-dependent cell-mediated cytotoxicity (ADCC) of tumor cells is utilized in the treatment of various cancers overexpressing unique antigens, such as neuroblastoma, breast cancer, B cell lymphoma, and others. NK cells also express a family of receptors called killer immunoglobulin-like receptors (KIRs), which regulate the function and response of NK cells toward target cells through their interaction with their cognate ligands that are expressed on tumor cells. Genetic polymorphisms in KIR and KIR-ligands, as well as FcγRs may influence NK cell responsiveness in conjunction with mAb immunotherapies. This review focuses on current therapeutic mAbs, different strategies to augment the anti-tumor efficacy of ADCC, and genotypic factors that may influence patient responses to antibody-dependent immunotherapies.
Collapse
Affiliation(s)
- Wei Wang
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, USA
| | - Amy K. Erbe
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, USA
| | - Jacquelyn A. Hank
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, USA
| | - Zachary S. Morris
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, USA
| | - Paul M. Sondel
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, USA
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
88
|
Keating SE, Ní Chorcora C, Dring MM, Stallings RL, O'Meara A, Gardiner CM. Increased frequencies of the killer immunoglobulin-like receptor genes KIR2DL2 and KIR2DS2 are associated with neuroblastoma. ACTA ACUST UNITED AC 2015. [PMID: 26202659 DOI: 10.1111/tan.12608] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Neuroblastoma is the most common extra-cranial solid tumour in children. Natural killer (NK) cells are innate lymphocytes that are known to mediate the direct cytotoxicity of neuroblastoma tumour cells. Natural variation in the highly polymorphic killer immunoglobulin-like receptors (KIR) and their cognate human leukocyte antigen (HLA) class I ligands results in considerable diversity in NK cell function. As the early onset of neuroblastoma suggests the contribution of genetic factors, we investigated if individual KIR genes, combined KIR gene haplotypes or compound KIR-HLA ligand genotypes could influence susceptibility to neuroblastoma. Genotype analysis of the KIR genes as well as their three major HLA class I ligand groups, HLA-C1, HLA-C2 and HLA-Bw4, was carried out in a cohort of 201 neuroblastoma patients compared with 240 healthy control subjects using polymerase chain reaction with sequence-specific primers. We found a significant increase in the frequency of KIR2DL2 (P = 0.019) as well as KIR2DS2 (P = 0.008) in patients with neuroblastoma compared with the healthy control group. While the incidence of the least inhibitory compound KIR-HLA-C genotype, KIR2DL3 in the presence of HLA-C1 was slightly reduced in neuroblastoma patients, this did not reach statistical significance (P = 0.069). In summary, while KIR-HLA compound genotypes have previously been implicated in predicting treatment outcomes in neuroblastoma, here we show that the presence of the individual KIR genes, KIR2DL2 and KIR2DS2, irrespective of HLA-C genotype is associated with the onset of this embryonal malignancy.
Collapse
Affiliation(s)
- S E Keating
- Natural Killer Cell Research Group, School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
| | - C Ní Chorcora
- Natural Killer Cell Research Group, School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
| | - M M Dring
- Natural Killer Cell Research Group, School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
| | - R L Stallings
- Department of Medicine and Therapeutics, The Royal College of Surgeons in Ireland, St. Stephen's Green, Dublin, Ireland
| | - A O'Meara
- National Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland
| | - C M Gardiner
- Natural Killer Cell Research Group, School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
| |
Collapse
|
89
|
Matlawska-Wasowska K, Gale JM, Nickl CK, Khalili P, Shirley B, Wilson BS, Vasef MA, Winter SS. Pyrosequencing for classification of human FcγRIIIA allotypes: a comparison with PCR-based techniques. Mol Diagn Ther 2015; 18:665-73. [PMID: 25230857 DOI: 10.1007/s40291-014-0120-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Surface-specific antigens expressed by hematopoietic cells are attractive targets for antibody-mediated immunotherapy. Monoclonal antibodies (mAbs) involve various mechanisms to eliminate target cells, including antibody-dependent cellular cytotoxicity (ADCC)- and phagocytosis (ADCP)-mediated killing through natural killer (NK) and macrophage effector cells bearing FcγRIIIA (CD16). The clinical efficacy of ADCC is particularly impacted by a single nucleotide polymorphism (SNP) found in the gene encoding FcγRIIIA (FCGR3A), which generates a variable distribution of the 158 V/V, F/V or F/F CD16 allotypes (F = phenylalanine, V = valine) in the normal human population. Currently, most patients are not screened for CD16 allotypes, creating the potential to include in their treatment a mAb-based therapy that may have limited benefit. Therefore, it is important to identify CD16 allotypes when considering mAb therapies that require ADCC/ADCP. OBJECTIVE The objective of this study was to develop a reliable PCR-based assay for classification of human FcγRIIIA allotypes. METHODS We studied 42 normal human subjects for the incidence of FcγRIIIA-158 polymorphisms using comparative molecular approaches. RESULTS The results of our study showed 100% accuracy in genotyping by pyrosequencing. In contrast, nested PCR-based allele-specific restriction assay and quantitative PCR techniques proved to be relatively less sensitive and less specific in distinguishing variant genotypes. CONCLUSION Since the efficacy of the mAb-based targeted immunotherapy may be highly dependent upon the CD16 polymorphism in a given individual, we recommend pyrosequencing for CD16 allotype testing.
Collapse
Affiliation(s)
- Ksenia Matlawska-Wasowska
- Department of Pediatrics, Division of Hematology and Oncology, MSC 10 5590, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA,
| | | | | | | | | | | | | | | |
Collapse
|
90
|
Abstract
Given recent technological advances and advances in our understanding of cancer, immunotherapy of cancer is being used with clear clinical benefit. The immunosuppression accompanying cancer itself, as well as with current cancer treatment with radiation or chemotherapy, impairs adaptive immune effectors to a greater extent than innate effector cells. In addition to being less suppressed, innate immune cells are capable of being enhanced via immune-stimulatory regimens. Most strategies being investigated to promote innate immune responses against cancer do not require complex, patient-specific, ex vivo cellular or molecular creation of therapeutic agents; thus they can, generally, be used as "off the shelf" therapeutics that could be administered by most cancer clinics. Successful applications of innate immunotherapy in the clinic have effectively targeted components of the innate immune response. Preclinical data demonstrate how initiation of innate immune responses can lead to subsequent adaptive long-term cancer immunity. We hypothesize that integration of innate immune activation strategies into combination therapies for cancer treatment will lead to more effective and long-term clinical benefit.
Collapse
Affiliation(s)
- Jacob L Goldberg
- Department of Pediatrics, The University of Wisconsin, Madison WI
| | - Paul M Sondel
- Department of Pediatrics, The University of Wisconsin, Madison WI; Department of Human Oncology, The University of Wisconsin, Madison WI; Department of Genetics, The University of Wisconsin, Madison WI.
| |
Collapse
|
91
|
Abstract
The past decade has seen several anticancer immunotherapeutic strategies transition from "promising preclinical models" to treatments with proven clinical activity or benefit. In 2013, the journal Science selected the field of Cancer Immunotherapy as the overall number-1 breakthrough for the year in all of scientific research. In the setting of cancer immunotherapy for adult malignancies, many of these immunotherapy strategies have relied on the cancer patient's endogenous antitumor T-cell response. Although much promising research in pediatric oncology is similarly focused on T-cell reactivity, several pediatric malignancies themselves, or the chemo-radiotherapy used to achieve initial responses, can be associated with profound immune suppression, particularly of the T-cell system. A separate component of the immune system, also able to mediate antitumor effects and less suppressed by conventional cancer treatment, is the NK-cell system. In recent years, several distinct immunotherapeutic approaches that rely on the activity of NK cells have moved from preclinical development into clinical testing, and some have shown clear antitumor benefit. This review provides an overview of NK cell-based immunotherapy efforts that are directed toward childhood malignancies, with an emphasis on protocols that are already in clinical testing.
Collapse
|
92
|
Abstract
Natural killer (NK) cells are normal white blood cells capable of killing malignant cells without prior sensitization. Allogeneic NK cell infusions are attractive for cancer therapy because of non-cross-resistant mechanisms of action and minimal overlapping toxicities with standard cancer treatments. Although NK therapy is promising, many obstacles will need to be overcome, including insufficient cell numbers, failure of homing to tumor sites, effector dysfunction, exhaustion, and tumor cell evasion. Capitalizing on the wealth of knowledge generated by recent NK cell biology studies and the advancements in biotechnology, substantial progress has been made recently in improving therapeutic efficiency and reducing side effects. A multipronged strategy is essential, including immunogenetic-based donor selection, refined NK cell bioprocessing, and novel augmentation techniques, to improve NK function and to reduce tumor resistance. Although data from clinical trials are currently limited primarily to hematologic malignancies, broader applications to a wide spectrum of adult and pediatric cancers are under way. The unique properties of human NK cells open up a new arena of novel cell-based immunotherapy against cancers that are resistant to contemporary therapies.
Collapse
Affiliation(s)
- Wing Leung
- Author's Affiliations: Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital; and Department of Pediatrics, University of Tennessee, Memphis, Tennessee
| |
Collapse
|
93
|
Suzuki M, Cheung NKV. Disialoganglioside GD2 as a therapeutic target for human diseases. Expert Opin Ther Targets 2015; 19:349-62. [PMID: 25604432 DOI: 10.1517/14728222.2014.986459] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Ganglioside GD2 is found in vertebrates and invertebrates, overexpressed among pediatric and adult solid tumors, including neuroblastoma, glioma, retinoblastoma, Ewing's family of tumors, rhabdomyosarcoma, osteosarcoma, leiomyosarcoma, liposarcoma, fibrosarcoma, small cell lung cancer and melanoma. It is also found on stem cells, neurons, some nerve fibers and basal layer of the skin. AREAS COVERED GD2 provides a promising clinical target for radiolabeled antibodies, bispecific antibodies, chimeric antigen receptor (CAR)-modified T cells, drug conjugates, nanoparticles and vaccines. Here, we review its biochemistry, normal physiology, role in tumorigenesis, important characteristics as a target, as well as anti-GD2-targeted strategies. EXPERT OPINION Bridging the knowledge gaps in understanding the interactions of GD2 with signaling molecules within the glycosynapses, and the regulation of its cellular expression should improve therapeutic strategies targeting this ganglioside. In addition to anti-GD2 IgG mAbs, their drug conjugates, radiolabeled forms especially when genetically engineered to improve therapeutic index and novel bispecific forms or CARs to retarget T-cells are promising candidates for treating metastatic cancers.
Collapse
Affiliation(s)
- Maya Suzuki
- Memorial Sloan Kettering Cancer Center, Department of Pediatrics , 1275 York Avenue, New York, NY 10065 , USA +1 646 888 2313 ; +1 631 422 0452 ;
| | | |
Collapse
|
94
|
McDermott DF, Cheng SC, Signoretti S, Margolin KA, Clark JI, Sosman JA, Dutcher JP, Logan TF, Curti BD, Ernstoff MS, Appleman L, Wong MKK, Khushalani NI, Oleksowicz L, Vaishampayan UN, Mier JW, Panka DJ, Bhatt RS, Bailey AS, Leibovich BC, Kwon ED, Kabbinavar FF, Belldegrun AS, Figlin RA, Pantuck AJ, Regan MM, Atkins MB. The high-dose aldesleukin "select" trial: a trial to prospectively validate predictive models of response to treatment in patients with metastatic renal cell carcinoma. Clin Cancer Res 2014; 21:561-8. [PMID: 25424850 DOI: 10.1158/1078-0432.ccr-14-1520] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
PURPOSE High-dose aldesleukin (HD IL2) received FDA approval for the treatment of metastatic renal cell carcinoma (MRCC) in 1992, producing a 14% objective response rate (ORR) and durable remissions. Retrospective studies suggested that clinical and pathologic features could predict for benefit. The Cytokine Working Group conducted this prospective trial to validate proposed predictive markers of response to HD IL2. EXPERIMENTAL DESIGN Standard HD IL2 was administered to prospectively evaluate whether the ORR of patients with mRCC with "good" predictive pathologic features based on an "integrated selection" model [ISM (e.g., clear-cell histology subclassification and carbonic anhydrase-9 (CA-9) IHC staining] was significantly higher than the ORR of a historical, unselected population. Archived tumor was collected for pathologic analysis including tumor programmed death-ligand 1 (PD-L1) expression. RESULTS One hundred and twenty eligible patients were enrolled between June 11 and September 7; 70% were Memorial Sloan Kettering Cancer Center (New York, NY) intermediate risk, 96% had clear cell RCC, and 99% had prior nephrectomy. The independently assessed ORR was 25% (30/120, 95% CI, 17.5%-33.7%, P = 0.0014; 3 complete responses, 27 partial responses) and was higher than a historical ORR. Thirteen patients (11%) remained progression free at 3 years and the median overall survival was 42.8 months. ORR was not statistically different by ISM classification ("good-risk" 23% vs. "poor-risk" 30%; P = 0.39). ORR was positively associated with tumor PD-L1 expression (P = 0.01) by IHC. CONCLUSIONS In this prospective, biomarker validation study, HD IL2 produced durable remissions and prolonged survival in both "good" and "poor-risk" patients. The proposed ISM was unable to improve the selection criteria. Novel markers (e.g., tumor PD L1 expression) appeared useful, but require independent validation.
Collapse
Affiliation(s)
- David F McDermott
- Beth Israel Deaconess Medical Center, Dana-Farber/Harvard Cancer Center, Boston, Massachusetts.
| | | | - Sabina Signoretti
- Brigham and Women's Hospital, Dana-Farber/Harvard Cancer Center, Boston, Massachusetts
| | | | - Joseph I Clark
- Loyola University, Stritch School of Medicine, Chicago, Illinois
| | - Jeffrey A Sosman
- Vanderbilt University Medical Center, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | | | | | - Brendan D Curti
- Providence Cancer Center, Earle A. Chiles Research Institute, Portland, Oregon
| | - Marc S Ernstoff
- Dartmouth-Hitchcock Medical Center, Norris Cotton Cancer Center, Dartmouth, New Hampshire
| | - Leonard Appleman
- Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | | | | | | | | | - James W Mier
- Beth Israel Deaconess Medical Center, Dana-Farber/Harvard Cancer Center, Boston, Massachusetts
| | - David J Panka
- Beth Israel Deaconess Medical Center, Dana-Farber/Harvard Cancer Center, Boston, Massachusetts
| | - Rupal S Bhatt
- Beth Israel Deaconess Medical Center, Dana-Farber/Harvard Cancer Center, Boston, Massachusetts
| | - Alexandra S Bailey
- Beth Israel Deaconess Medical Center, Dana-Farber/Harvard Cancer Center, Boston, Massachusetts
| | | | - Eugene D Kwon
- Department of Urology, Mayo Clinic, Rochester, Minnesota
| | | | - Arie S Belldegrun
- University of California Los Angeles Medical Center, Santa Monica, California
| | - Robert A Figlin
- Cedars-Sinai's Samuel Oschin Comprehensive Cancer Institute, West Hollywood, California
| | - Allan J Pantuck
- University of California Los Angeles Medical Center, Santa Monica, California
| | | | | |
Collapse
|
95
|
Abstract
After decades of research, immunotherapies for cancer are demonstrating increasing success. These agents can amplify existent antitumour immunity or induce durable antitumour immune responses in a wide array of cancers. The spectrum of immunotherapeutics is broad, spanning monoclonal antibodies and their derivatives, tumour vaccines, and adoptive therapies using T cells and natural killer cells. Only a small number of immunotherapies have been tested in paediatric cancers, but impressive antitumour effects have already been observed. Mononclonal antibodies targeting GD2 that induce antibody-dependent cell-mediated cytotoxicity improve survival in high-risk neuroblastoma. Bi-specific monoclonal antibodies that simultaneously target CD19 and activate T cells can induce remission in acute B-cell lymphoblastic leukaemia (B-ALL) and adoptive immunotherapy using T cells genetically engineered to express chimeric antigen receptors targeting CD19 induce impressive responses in B-ALL. Efforts are underway to generate and test new immunotherapies in a wider array of paediatric cancers. Major challenges include a need to identify immunotherapy targets on the most lethal childhood cancers, to expand availability of technology-intense platforms, such as adoptive cell therapy, to optimize management of novel toxicities associated with this new class of cancer therapies and to determine how best to incorporate these therapies into standard treatment paradigms.
Collapse
|
96
|
Skrombolas D, Frelinger JG. Challenges and developing solutions for increasing the benefits of IL-2 treatment in tumor therapy. Expert Rev Clin Immunol 2014; 10:207-17. [PMID: 24410537 DOI: 10.1586/1744666x.2014.875856] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Interleukin-2 (IL-2) is a cytokine with pleiotropic effects on the immune system. Systemic IL-2 treatment has produced durable responses in melanoma and renal cancer patients, but unfortunately this is effective only in a fraction of patients. Moreover, IL-2 treatment also engenders serious side effects, which limit its clinical utility. It is now appreciated that IL-2 not only stimulates NK and effector T cells but also has a critical role in the generation and maintenance of regulatory T cells, which act to dampen immune responses. Thus, successful immunotherapy of cancers using IL-2 has to address two fundamentally important issues: (1) how to limit side effects yet be active where it is needed, and (2) how to preferentially activate effector T cells while limiting the stimulation of Tregs. Strategies are now being developed to address these critical obstacles that may lead to a renaissance of IL-2 therapy.
Collapse
Affiliation(s)
- Denise Skrombolas
- Department of Microbiology and Immunology, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY14642, USA
| | | |
Collapse
|
97
|
Du J, Lopez-Verges S, Pitcher BN, Johnson J, Jung SH, Zhou L, Hsu K, Czuczman MS, Cheson B, Kaplan L, Lanier LL, Venstrom JM. CALGB 150905 (Alliance): rituximab broadens the antilymphoma response by activating unlicensed NK cells. Cancer Immunol Res 2014; 2:878-89. [PMID: 24958280 PMCID: PMC4264658 DOI: 10.1158/2326-6066.cir-13-0158] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Natural killer (NK) cells contribute to clinical responses in patients treated with rituximab, but the rules determining NK-cell responsiveness to mAb therapies are poorly defined. A deeper understanding of the mechanisms responsible for antibody-dependent cellular cytotoxicity (ADCC) could yield useful biomarkers for predicting clinical responses in patients. Unlicensed NK cells, defined as NK cells lacking expression of an inhibitory KIR for self-HLA class I ligands, are hyporesponsive in steady state, but are potent effectors in inflammatory conditions. We hypothesized that antitumor antibodies such as rituximab can overcome NK-cell dependence on licensing, making unlicensed NK cells important for clinical responses. Here, we examined the influences of variations in KIR and HLA class I alleles on in vitro responses to rituximab. We tested the clinical significance in a cohort of patients with follicular lymphoma treated with rituximab-containing mAb combinations, and show that rituximab triggers responses from all NK-cell populations regardless of licensing. Neither IL2 nor accessory cells are required for activating unlicensed NK cells, but both can augment rituximab-mediated ADCC. Moreover, in 101 patients with follicular lymphoma treated with rituximab-containing mAb combinations, a "missing ligand" genotype (predictive of unlicensed NK cells) is associated with a higher rate of progression-free survival. Our data suggest that the clinical efficacy of rituximab may be driven, in part, by its ability to broaden the NK-cell repertoire to include previously hyporesponsive, unlicensed NK cells. A "missing ligand" KIR and HLA class I genotype may be predictive of this benefit and useful for personalizing treatment decisions in lymphomas and other tumors.
Collapse
Affiliation(s)
- Juan Du
- Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Sandra Lopez-Verges
- Department of Microbiology and Immunology and the Cancer Research Institute, University of California, San Francisco, San Francisco, California
| | - Brandelyn N Pitcher
- Alliance Statistics and Data Center, Duke University Medical Center, Durham, North Carolina
| | - Jeffrey Johnson
- Alliance Statistics and Data Center, Duke University Medical Center, Durham, North Carolina
| | - Sin-Ho Jung
- Alliance Statistics and Data Center, Duke University Medical Center, Durham, North Carolina
| | - Lili Zhou
- Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Katharine Hsu
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Myron S Czuczman
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York
| | - Bruce Cheson
- Department of Medicine, Georgetown University, Washington, District of Columbia
| | - Lawrence Kaplan
- Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Lewis L Lanier
- Department of Microbiology and Immunology and the Cancer Research Institute, University of California, San Francisco, San Francisco, California
| | - Jeffrey M Venstrom
- Department of Medicine, University of California, San Francisco, San Francisco, California.
| |
Collapse
|
98
|
Abstract
Ganglioside GD2 is a tumor-associated surface antigen found in a broad spectrum of human cancers and stem cells. They include pediatric embryonal tumors (neuroblastoma, retinoblastoma, brain tumors, osteosarcoma, Ewing sarcoma, rhabdomyosarcoma), as well as adult cancers (small cell lung cancer, melanoma, soft tissue sarcomas). Because of its restricted normal tissue distribution, GD2 has been proven safe for antibody targeting. Anti-GD2 antibody is now incorporated into the standard of care for the treatment of high-risk metastatic neuroblastoma. Building on this experience, novel combinations of antibodies, cytokines, cells, and genetically engineered products all directed at GD2 are rapidly moving into the clinic. In this review, past and present immunotherapy trials directed at GD2 will be summarized, highlighting the lessons learned and the future directions.
Collapse
Affiliation(s)
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY.
| |
Collapse
|
99
|
Cheng M, Ahmed M, Xu H, Cheung NKV. Structural design of disialoganglioside GD2 and CD3-bispecific antibodies to redirect T cells for tumor therapy. Int J Cancer 2014; 136:476-86. [PMID: 24895182 DOI: 10.1002/ijc.29007] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 04/21/2014] [Accepted: 05/08/2014] [Indexed: 02/02/2023]
Abstract
Antibody-based immunotherapy has proven efficacy for patients with high-risk neuroblastoma. However, despite being the most efficient tumoricidal effectors, T cells are underutilized because they lack Fc receptors. Using a monovalent single-chain fragment (ScFv) platform, we engineered tandem scFv bispecific antibodies (BsAbs) that specifically target disialoganglioside (GD2) on tumor cells and CD3 on T cells. Structural variants of BsAbs were constructed and ranked based on binding to GD2, and on competency in inducing T-cell-mediated tumor cytotoxicity. In vitro thermal stability and binding measurements were used to characterize each of the constructs, and in silico molecular modeling was used to show how the orientation of the variable region heavy (VH) and light (VL) chains of the anti-GD2 ScFv could alter the conformations of key residues responsible for high affinity binding. We showed that the VH-VL orientation, the (GGGGS)3 linker, disulfide bond stabilization of scFv, when combined with an affinity matured mutation provided the most efficient BsAb to direct T cells to lyse GD2-positive tumor cells. In vivo, the optimized BsAb could efficiently inhibit melanoma and neuroblastoma xenograft growth. These findings provide preclinical validation of a structure-based method to assist in designing BsAb for T-cell-mediated therapy.
Collapse
Affiliation(s)
- Ming Cheng
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | | | | |
Collapse
|
100
|
Terszowski G, Klein C, Stern M. KIR/HLA interactions negatively affect rituximab- but not GA101 (obinutuzumab)-induced antibody-dependent cellular cytotoxicity. THE JOURNAL OF IMMUNOLOGY 2014; 192:5618-24. [PMID: 24795454 DOI: 10.4049/jimmunol.1400288] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Ab-dependent cellular cytotoxicity (ADCC) mediated by NK cells is regulated by inhibitory killer cell Ig-like receptors (KIRs), which interact with target cell HLA class I. We analyzed how KIR/HLA interactions influence ADCC induced by rituximab and by GA101, a novel type II CD20 Ab glycoengineered for increased FcgRIII binding and ADCC capacity. We found that KIR/HLA interactions strongly and selectively inhibit rituximab-induced in vitro ADCC toward target cells expressing cognate HLA KIR ligands. NK cells of donors carrying all three ligands to inhibitory KIR showed weak activation and target cell depletion capacity when incubated with rituximab and KIR-ligand matched target B cells. In contrast, NK cells from individuals missing one or more KIR ligands activated more strongly and depleted KIR ligand-matched target B cells more efficiently in the presence of rituximab. NK cells expressing a KIR for which the ligand was absent were the main effectors of ADCC in these donors. Notably, the influence of KIR/HLA interactions on NK cell activation was synergistic with the effect of the V158F FCGR3A single nucleotide polymorphism. In contrast, GA101 induced activation of NK cells irrespective of inhibitory KIR expression, and efficiency of target cell depletion was not negatively affected by KIR/HLA interactions. These data show that modification of the Fc fragment to enhance ADCC can be an effective strategy to augment the efficacy of therapeutic mAbs by recruiting NK cells irrespective of their inhibitory KIR expression.
Collapse
Affiliation(s)
- Grzegorz Terszowski
- Department of Biomedicine, University Hospital Basel, CH-4031 Basel, Switzerland; and
| | - Christian Klein
- Pharma Research and Early Development, Roche Glycart AG, CH-8952 Schlieren, Switzerland
| | - Martin Stern
- Department of Biomedicine, University Hospital Basel, CH-4031 Basel, Switzerland; and
| |
Collapse
|