1
|
Frampton S, Smith R, Ferson L, Gibson J, Hollox EJ, Cragg MS, Strefford JC. Fc gamma receptors: Their evolution, genomic architecture, genetic variation, and impact on human disease. Immunol Rev 2024. [PMID: 39345014 DOI: 10.1111/imr.13401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Fc gamma receptors (FcγRs) are a family of receptors that bind IgG antibodies and interface at the junction of humoral and innate immunity. Precise regulation of receptor expression provides the necessary balance to achieve healthy immune homeostasis by establishing an appropriate immune threshold to limit autoimmunity but respond effectively to infection. The underlying genetics of the FCGR gene family are central to achieving this immune threshold by regulating affinity for IgG, signaling efficacy, and receptor expression. The FCGR gene locus was duplicated during evolution, retaining very high homology and resulting in a genomic region that is technically difficult to study. Here, we review the recent evolution of the gene family in mammals, its complexity and variation through copy number variation and single-nucleotide polymorphism, and impact of these on disease incidence, resolution, and therapeutic antibody efficacy. We also discuss the progress and limitations of current approaches to study the region and emphasize how new genomics technologies will likely resolve much of the current confusion in the field. This will lead to definitive conclusions on the impact of genetic variation within the FCGR gene locus on immune function and disease.
Collapse
Affiliation(s)
- Sarah Frampton
- Cancer Genomics Group, Faculty of Medicine, School of Cancer Sciences, University of Southampton, Southampton, UK
| | - Rosanna Smith
- Antibody and Vaccine Group, Faculty of Medicine, School of Cancer Sciences, Centre for Cancer Immunology, University of Southampton, Southampton, UK
| | - Lili Ferson
- Cancer Genomics Group, Faculty of Medicine, School of Cancer Sciences, University of Southampton, Southampton, UK
| | - Jane Gibson
- Cancer Genomics Group, Faculty of Medicine, School of Cancer Sciences, University of Southampton, Southampton, UK
| | - Edward J Hollox
- Department of Genetics, Genomics and Cancer Sciences, College of Life Sciences, University of Leicester, Leicester, UK
| | - Mark S Cragg
- Antibody and Vaccine Group, Faculty of Medicine, School of Cancer Sciences, Centre for Cancer Immunology, University of Southampton, Southampton, UK
| | - Jonathan C Strefford
- Cancer Genomics Group, Faculty of Medicine, School of Cancer Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
2
|
Hong SK, Lee KW, Kim JY, Lee J, Kim J, Choi HH, Hong SY, Lee JM, Choi Y, Yi NJ, Suh KS. Factors associated with rituximab-mediated B cell depletion in ABO-incompatible adult living donor liver transplantation. KOREAN JOURNAL OF TRANSPLANTATION 2023; 37:170-178. [PMID: 37694598 PMCID: PMC10583967 DOI: 10.4285/kjt.23.0031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/06/2023] [Accepted: 07/13/2023] [Indexed: 09/12/2023] Open
Abstract
Background Pretransplant therapies such as rituximab and plasmapheresis have led to an increase in ABO-incompatible (ABOi) living donor liver transplantation (LDLT), thus helping to overcome organ shortages. This study evaluated the changes in anti-A/B titers and CD19 levels over time in patients undergoing ABOi LT and aimed to understand the effect of single-nucleotide polymorphisms (SNPs) in Fc gamma receptor (FcγR) on rituximab therapy. Methods Two SNPs of FCGR2A (131H/R) and FCGR3A (158F/V) were identified. The clinical data on 44 patients who underwent ABOi LDLT between May 2019 and October 2021 at Seoul National University Hospital were reviewed retrospectively. Results Following desensitization with rituximab and subsequent LDLT, the anti-A/B titer recovered within 1 week, but decreased thereafter. The CD19 level increased at 3 months after LT. The genotyping data for FCGR3A (158F/V) indicated that two patients had the V/V genotype, and 42 had the F/V genotype. In the genotyping data for FCGR2A (131H/R), 21 patients had the H/H genotype, three had the R/R genotype, and 20 had the H/R genotype. However, there were no significant differences in anti-A/B and CD19 levels, bacteremia rates, T cell-mediated rejection, antibody-mediated rejection, or the survival rate among the FCGR2A types. Conclusions There were significant changes in the anti-A/B titers and CD19 levels over time in each patient after ABOi LDLT. The difference in outcomes following LT according to the FcγR SNP type for rituximab was unclear. Further studies with larger sample sizes are needed to confirm the effect of FcγR SNPs on rituximab therapy.
Collapse
Affiliation(s)
- Suk Kyun Hong
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Kwang-Woong Lee
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Jae-Yoon Kim
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Jaewon Lee
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Jiyoung Kim
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Hyun Hwa Choi
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Su young Hong
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Jeong-Moo Lee
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - YoungRok Choi
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Nam-Joon Yi
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Kyung-Suk Suh
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
3
|
Gonzalez JC, Chakraborty S, Thulin NK, Wang TT. Heterogeneity in IgG-CD16 signaling in infectious disease outcomes. Immunol Rev 2022; 309:64-74. [PMID: 35781671 PMCID: PMC9539944 DOI: 10.1111/imr.13109] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
In this review, we discuss how IgG antibodies can modulate inflammatory signaling during viral infections with a focus on CD16a-mediated functions. We describe the structural heterogeneity of IgG antibody ligands, including subclass and glycosylation that impact binding by and downstream activity of CD16a, as well as the heterogeneity of CD16a itself, including allele and expression density. While inflammation is a mechanism required for immune homeostasis and resolution of acute infections, we focus here on two infectious diseases that are driven by pathogenic inflammatory responses during infection. Specifically, we review and discuss the evolving body of literature showing that afucosylated IgG immune complex signaling through CD16a contributes to the overwhelming inflammatory response that is central to the pathogenesis of severe forms of dengue disease and coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
- Joseph C. Gonzalez
- Department of Medicine, Division of Infectious DiseasesStanford University School of MedicineStanfordCaliforniaUSA,Program in ImmunologyStanford University School of MedicineStanfordCaliforniaUSA
| | - Saborni Chakraborty
- Department of Medicine, Division of Infectious DiseasesStanford University School of MedicineStanfordCaliforniaUSA
| | - Natalie K. Thulin
- Department of ImmunologyUniversity of WashingtonSeattleWashingtonUSA
| | - Taia T. Wang
- Department of Medicine, Division of Infectious DiseasesStanford University School of MedicineStanfordCaliforniaUSA,Program in ImmunologyStanford University School of MedicineStanfordCaliforniaUSA,Department of Microbiology and ImmunologyStanford University School of MedicineStanfordCaliforniaUSA
| |
Collapse
|
4
|
Mata-Molanes JJ, Rebollo-Liceaga J, Martínez-Navarro EM, Manzano RG, Brugarolas A, Juan M, Sureda M. Relevance of Fc Gamma Receptor Polymorphisms in Cancer Therapy With Monoclonal Antibodies. Front Oncol 2022; 12:926289. [PMID: 35814459 PMCID: PMC9263556 DOI: 10.3389/fonc.2022.926289] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/30/2022] [Indexed: 12/15/2022] Open
Abstract
Therapeutic monoclonal antibodies (mAbs), including immune checkpoint inhibitors (ICIs), are an important breakthrough for the treatment of cancer and have dramatically changed clinical outcomes in a wide variety of tumours. However, clinical response varies among patients receiving mAb-based treatment, so it is necessary to search for predictive biomarkers of response to identify the patients who will derive the greatest therapeutic benefit. The interaction of mAbs with Fc gamma receptors (FcγR) expressed by innate immune cells is essential for antibody-dependent cellular cytotoxicity (ADCC) and this binding is often critical for their in vivo efficacy. FcγRIIa (H131R) and FcγRIIIa (V158F) polymorphisms have been reported to correlate with response to therapeutic mAbs. These polymorphisms play a major role in the affinity of mAb receptors and, therefore, can exert a profound impact on antitumor response in these therapies. Furthermore, recent reports have revealed potential mechanisms of ICIs to modulate myeloid subset composition within the tumour microenvironment through FcγR-binding, optimizing their anti-tumour activity. The purpose of this review is to highlight the clinical contribution of FcγR polymorphisms to predict response to mAbs in cancer patients.
Collapse
Affiliation(s)
- Juan J. Mata-Molanes
- Oncology Platform, Hospital Quirónsalud Torrevieja, Alicante, Spain
- *Correspondence: Juan J. Mata-Molanes,
| | | | | | | | | | - Manel Juan
- Department of Immunology, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Manuel Sureda
- Oncology Platform, Hospital Quirónsalud Torrevieja, Alicante, Spain
| |
Collapse
|
5
|
Kusowska A, Kubacz M, Krawczyk M, Slusarczyk A, Winiarska M, Bobrowicz M. Molecular Aspects of Resistance to Immunotherapies-Advances in Understanding and Management of Diffuse Large B-Cell Lymphoma. Int J Mol Sci 2022; 23:ijms23031501. [PMID: 35163421 PMCID: PMC8835809 DOI: 10.3390/ijms23031501] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/22/2022] [Accepted: 01/26/2022] [Indexed: 12/28/2022] Open
Abstract
Despite the unquestionable success achieved by rituximab-based regimens in the management of diffuse large B-cell lymphoma (DLBCL), the high incidence of relapsed/refractory disease still remains a challenge. The widespread clinical use of chemo-immunotherapy demonstrated that it invariably leads to the induction of resistance; however, the molecular mechanisms underlying this phenomenon remain unclear. Rituximab-mediated therapeutic effect primarily relies on complement-dependent cytotoxicity and antibody-dependent cell cytotoxicity, and their outcome is often compromised following the development of resistance. Factors involved include inherent genetic characteristics and rituximab-induced changes in effectors cells, the role of ligand/receptor interactions between target and effector cells, and the tumor microenvironment. This review focuses on summarizing the emerging advances in the understanding of the molecular basis responsible for the resistance induced by various forms of immunotherapy used in DLBCL. We outline available models of resistance and delineate solutions that may improve the efficacy of standard therapeutic protocols, which might be essential for the rational design of novel therapeutic regimens.
Collapse
Affiliation(s)
- Aleksandra Kusowska
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (A.K.); (M.K.); (M.K.); (A.S.); (M.W.)
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Matylda Kubacz
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (A.K.); (M.K.); (M.K.); (A.S.); (M.W.)
| | - Marta Krawczyk
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (A.K.); (M.K.); (M.K.); (A.S.); (M.W.)
- Laboratory of Immunology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
- Doctoral School of Translational Medicine, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland
| | - Aleksander Slusarczyk
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (A.K.); (M.K.); (M.K.); (A.S.); (M.W.)
- Department of General, Oncological and Functional Urology, Medical University of Warsaw, 02-005 Warsaw, Poland
| | - Magdalena Winiarska
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (A.K.); (M.K.); (M.K.); (A.S.); (M.W.)
- Laboratory of Immunology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Malgorzata Bobrowicz
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (A.K.); (M.K.); (M.K.); (A.S.); (M.W.)
- Correspondence:
| |
Collapse
|
6
|
Lavacchi D, Landini I, Perrone G, Roviello G, Mini E, Nobili S. Pharmacogenetics in diffuse large B-cell lymphoma treated with R-CHOP: Still an unmet challenge. Pharmacol Ther 2021; 229:107924. [PMID: 34175369 DOI: 10.1016/j.pharmthera.2021.107924] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/19/2021] [Accepted: 05/24/2021] [Indexed: 02/07/2023]
Abstract
DLBCL is the most common lymphoma representing approximately one third of all non-Hodgkin lymphomas and about 40% of patients do not benefit of the standard first-line immune-chemotherapeutic treatment (i.e., R-CHOP - rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone) that is administered as upfront therapy to substantially all patients independently from the stage of disease and other prognostic parameters. The administration of other pharmacological treatments is in fact limited to selected patients, unfitting for R-CHOP. Although clinical prognostic scores, i.e. International Prognostic Index (IPI), and molecular classifiers based on the cell of origin are available, at present no biomarkers predictive of R-CHOP response has been identified and validated. Constitutional polymorphisms of genes involved in the mechanism of action of drugs included in R-CHOP have been suggested by many authors to play a role in the efficacy and in some case in the toxicity of this treatment. Thus, it is conceivable that in the future, after proper validation, some polymorphisms can be used as pharmacogenetic biomarkers of therapeutic outcome in this disease setting. This review discusses the status of the art on molecular biomarkers predictive of DLBCL prognosis and deals with the relevant issue of the variability in response to DLBCL drug treatment. Overall, this review focuses on single nucleotide polymorphisms (SNPs) that, based on a candidate gene approach or on a GWAS analysis, have been suggested to play a role in response to R-CHOP. In particular, SNPs discovered by a candidate gene approach are related to gene involved in drug transport (i.e. ATP-binding cassette transporters), drug metabolism, drug detoxification enzymes, oxidative stress, apoptosis, DNA repair, immunity and angiogenesis. Data from a GWAS analysis performed in DLBCL patients treated with R-CHOP, identified two SNPs associated with clinical outcomes related to genes involved in pivotal cellular processes and in transcriptional regulation and cell cycle progression, respectively. Ongoing prospective pharmacogenetic clinical trials, including a GWAS study we performed, have also been discussed.
Collapse
Affiliation(s)
- Daniele Lavacchi
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy
| | - Ida Landini
- Department of Health Sciences, University of Florence, Florence, Italy; DENOTHE Excellence Center, University of Florence, Florence, Italy
| | - Gabriele Perrone
- Department of Health Sciences, University of Florence, Florence, Italy; DENOTHE Excellence Center, University of Florence, Florence, Italy
| | - Giandomenico Roviello
- Department of Health Sciences, University of Florence, Florence, Italy; DENOTHE Excellence Center, University of Florence, Florence, Italy
| | - Enrico Mini
- Department of Health Sciences, University of Florence, Florence, Italy; DENOTHE Excellence Center, University of Florence, Florence, Italy; Cancer Pharmacology Working Group of the Italian Society of Pharmacology, Milan, Italy.
| | - Stefania Nobili
- Cancer Pharmacology Working Group of the Italian Society of Pharmacology, Milan, Italy; Department of Neurosciences, Imaging and Clinical Sciences, "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy.
| |
Collapse
|
7
|
Vukovic N, van Elsas A, Verbeek JS, Zaiss DMW. Isotype selection for antibody-based cancer therapy. Clin Exp Immunol 2021; 203:351-365. [PMID: 33155272 PMCID: PMC7874837 DOI: 10.1111/cei.13545] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 10/16/2020] [Accepted: 10/29/2020] [Indexed: 01/14/2023] Open
Abstract
The clinical application of monoclonal antibodies (mAbs) has revolutionized the field of cancer therapy, as it has enabled the successful treatment of previously untreatable types of cancer. Different mechanisms play a role in the anti-tumour effect of mAbs. These include blocking of tumour-specific growth factor receptors or of immune modulatory molecules as well as complement and cell-mediated tumour cell lysis. Thus, for many mAbs, Fc-mediated effector functions critically contribute to the efficacy of treatment. As immunoglobulin (Ig) isotypes differ in their ability to bind to Fc receptors on immune cells as well as in their ability to activate complement, they differ in the immune responses they activate. Therefore, the choice of antibody isotype for therapeutic mAbs is dictated by its intended mechanism of action. Considering that clinical efficacy of many mAbs is currently achieved only in subsets of patients, optimal isotype selection and Fc optimization during antibody development may represent an important step towards improved patient outcome. Here, we discuss the current knowledge of the therapeutic effector functions of different isotypes and Fc-engineering strategies to improve mAbs application.
Collapse
Affiliation(s)
- N. Vukovic
- Institute of Immunology and Infection ResearchSchool of Biological SciencesUniversity of EdinburghAshworth LaboratoriesEdinburghUK
| | | | - J. S. Verbeek
- Department of Biomedical EngineeringToin University of YokohamaYokohamaJapan
| | - D. M. W. Zaiss
- Institute of Immunology and Infection ResearchSchool of Biological SciencesUniversity of EdinburghAshworth LaboratoriesEdinburghUK
| |
Collapse
|
8
|
How gene polymorphisms can influence clinical response and toxicity following R-CHOP therapy in patients with diffuse large B cell lymphoma. Blood Rev 2017; 31:235-249. [DOI: 10.1016/j.blre.2017.02.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 12/07/2016] [Accepted: 02/03/2017] [Indexed: 12/20/2022]
|
9
|
Ogura M, Coiffier B, Kwon HC, Yoon SW. Scientific rationale for extrapolation of biosimilar data across cancer indications: case study of CT-P10. Future Oncol 2017; 13:45-53. [DOI: 10.2217/fon-2017-0156] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
For a biosimilar to gain regulatory approval, a comprehensive comparability exercise must demonstrate that it is highly similar to its originator biologic, or reference product. Once biosimilarity has been shown, it is possible to approve the biosimilar for additional indications held by the reference product, without clinical trials in these indications. Extrapolation of clinical data is permitted by regulatory agencies as long as it is scientifically justified. CT-P10, a biosimilar of rituximab, was recently approved in Europe for all indications held by its reference product, incorporating both autoimmune diseases and hematological cancers. Here, we review the scientific rationale for extrapolation in biosimilar development using the example of CT-P10 as a case study.
Collapse
Affiliation(s)
- Michinori Ogura
- Department of Hematology, Tokai Central Hospital, Gifu, Japan
| | | | - Hyuk-Chan Kwon
- CELLTRION Healthcare Co. Ltd, Incheon, Republic of Korea
| | - Sang Wook Yoon
- CELLTRION Healthcare Co. Ltd, Incheon, Republic of Korea
| |
Collapse
|
10
|
Brown EP, Dowell KG, Boesch AW, Normandin E, Mahan AE, Chu T, Barouch DH, Bailey-Kellogg C, Alter G, Ackerman ME. Multiplexed Fc array for evaluation of antigen-specific antibody effector profiles. J Immunol Methods 2017; 443:33-44. [PMID: 28163018 PMCID: PMC5333794 DOI: 10.1016/j.jim.2017.01.010] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 01/27/2017] [Accepted: 01/31/2017] [Indexed: 01/09/2023]
Abstract
Antibodies are widely considered to be a frequent primary and often mechanistic correlate of protection of approved vaccines; thus evaluating the antibody response is of critical importance in attempting to understand and predict the efficacy of novel vaccine candidates. Historically, antibody responses have been analyzed by determining the titer of the humoral response using measurements such as an ELISA, neutralization, or agglutination assays. In the simplest case, sufficiently high titers of antibody against vaccine antigen(s) are sufficient to predict protection. However, antibody titer provides only a partial measure of antibody function, which is dependent on both the variable region (Fv) to bind the antigen target, and the constant region (Fc) to elicit an effector response from the innate arm of the immune system. In the case of some diseases, such as HIV, for which an effective vaccine has proven elusive, antibody effector function has been shown to be an important driver of monoclonal antibody therapy outcomes, of viral control in infected patients, and of vaccine-mediated protection in preclinical and clinical studies. We sought to establish a platform for the evaluation of the Fc domain characteristics of antigen-specific antibodies present in polyclonal samples in order to better develop insights into Fc receptor-mediated antibody effector activity, more fully understand how antibody responses may differ in association with disease progression and between subject groups, and differentiate protective from non-protective responses. To this end we have developed a high throughput biophysical platform capable of simultaneously evaluating many dimensions of the antibody effector response. High-throughput array-based characterization platform for polyclonal antibodies. Development of a biophysical proxy for antibody effector function. Antigen and Fc receptor recognition characteristics are captured. Enables systematic serologic studies of NHP and human antibody samples.
Collapse
Affiliation(s)
- Eric P Brown
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Karen G Dowell
- Department of Computer Science, Dartmouth College, Hanover, NH 03755, USA
| | - Austin W Boesch
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Erica Normandin
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Alison E Mahan
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Thach Chu
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Dan H Barouch
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | - Galit Alter
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | | |
Collapse
|
11
|
The FCGR3A polymorphism predicts the response to rituximab-based therapy in patients with non-Hodgkin lymphoma: a meta-analysis. Ann Hematol 2016; 95:1483-90. [PMID: 27431582 DOI: 10.1007/s00277-016-2723-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Accepted: 06/03/2016] [Indexed: 12/16/2022]
Abstract
Epidemiological studies have assessed the association between Fc gamma receptor IIIA (FCGR3A) 158 V/F and the response to rituximab-based therapy in patients with non-Hodgkin lymphoma (NHL), but the findings have been inconsistent. We performed this meta-analysis to obtain a better assessment of this relationship. Electronic database searches were conducted for relevant studies. A pooled odds ratio (OR) with a 95 % confidence interval (95 % CI) was used to assess the strength of the association. Analyses of the subgroup and publication bias were conducted. A total of 10 studies involving 1050 patients were analyzed. In all the genetic models, no clear relationship was found between the FCGR3A 158 V/F polymorphism and the response to rituximab-based therapy in NHL patients. When categorized by ethnicity, Asian individuals with the FCGR3A 158 V/V allele (OR = 4.37; 95 % CI = 1.07-17.73; P = 0.039) or the non-F/(FV + VV) (OR = 2.50; 95 % CI = 1.04-5.98; P = 0.040) allele have a significantly higher complete response rate (CR) compared to FF individuals. No obvious heterogeneities were observed. In addition, no statistical evidence for a publication bias was found. Our study suggested that the FCGR3A 158 V/F polymorphism can predict the treatment response to rituximab-based chemotherapy in NHL patients, especially for Asian individuals.
Collapse
|
12
|
Abstract
Monoclonal antibody (mAb) immunotherapy is currently experiencing an unprecedented amount of success, delivering blockbuster sales for the pharmaceutical industry. Having experienced several false dawns and overcoming technical issues which limited progress, we are now entering a golden period where mAbs are becoming a mainstay of treatment regimes for diseases ranging from cancer to autoimmunity. In this review, we discuss how these mAbs are most likely working and focus in particular on the key receptors that they interact with to precipitate their therapeutic effects. Although their targets may vary, their engagement with Fcγ receptors (FcγRs) on numerous immune effector cells is almost universal, and here we review their roles in delivering successful immunotherapy.
Collapse
Affiliation(s)
- Lekh N Dahal
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, General Hospital, Southampton, UK
| | - Ali Roghanian
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, General Hospital, Southampton, UK
| | - Stephen A Beers
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, General Hospital, Southampton, UK
| | - Mark S Cragg
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, General Hospital, Southampton, UK
| |
Collapse
|
13
|
Hargreaves CE, Rose-Zerilli MJJ, Machado LR, Iriyama C, Hollox EJ, Cragg MS, Strefford JC. Fcγ receptors: genetic variation, function, and disease. Immunol Rev 2015; 268:6-24. [DOI: 10.1111/imr.12341] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- Chantal E. Hargreaves
- Cancer Genomics Group; Cancer Sciences; Faculty of Medicine; University of Southampton; Southampton UK
- Antibody and Vaccine Group; Cancer Sciences; Faculty of Medicine; University of Southampton; Southampton UK
| | | | - Lee R. Machado
- Department of Genetics; University of Leicester; Leicester UK
- School of Health; University of Northampton; Northampton UK
| | - Chisako Iriyama
- Department of Hematology and Oncology; Nagoya University Graduate School of Medicine; Nagoya Japan
| | | | - Mark S. Cragg
- Antibody and Vaccine Group; Cancer Sciences; Faculty of Medicine; University of Southampton; Southampton UK
| | - Jonathan C. Strefford
- Cancer Genomics Group; Cancer Sciences; Faculty of Medicine; University of Southampton; Southampton UK
| |
Collapse
|
14
|
Nielsen KR, Steffensen R, Haunstrup TM, Bødker JS, Dybkær K, Baech J, Bøgsted M, Johnsen HE. Inherited variation in immune response genes in follicular lymphoma and diffuse large B-cell lymphoma. Leuk Lymphoma 2015; 56:3257-66. [PMID: 26044172 DOI: 10.3109/10428194.2015.1058936] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Diffuse large B-cell lymphoma (DLBCL) and follicular lymphoma (FL) both depend on immune-mediated survival and proliferation signals from the tumor microenvironment. Inherited genetic variation influences this complex interaction. A total of 89 studies investigating immune-response genes in DLBCL and FL were critically reviewed. Relatively consistent association exists for variation in the tumor necrosis factor alpha (TNFA) and interleukin-10 loci and DLBCL risk; for DLBCL outcome association with the TNFA locus exists. Variations at chromosome 6p31-32 were associated with FL risk. Importantly, individual risk alleles have been shown to interact with each other. We suggest that the pathogenetic impact of polymorphic genes should include gene-gene interaction analysis and should be validated in preclinical model systems of normal B lymphopoiesis and B-cell malignancies. In the future, large cohort studies of interactions and genome-wide association studies are needed to extend the present findings and explore new risk alleles to be studied in preclinical models.
Collapse
Affiliation(s)
| | - Rudi Steffensen
- a Department of Clinical Immunology , Aalborg University Hospital , Denmark
| | | | | | - Karen Dybkær
- b Department of Haematology , Aalborg University Hospital.,c Clinical Cancer Research Center, Aalborg University Hospital , Denmark and Department of Clinical Medicine , Aalborg University , Denmark
| | - John Baech
- a Department of Clinical Immunology , Aalborg University Hospital , Denmark
| | - Martin Bøgsted
- b Department of Haematology , Aalborg University Hospital.,c Clinical Cancer Research Center, Aalborg University Hospital , Denmark and Department of Clinical Medicine , Aalborg University , Denmark
| | - Hans Erik Johnsen
- b Department of Haematology , Aalborg University Hospital.,c Clinical Cancer Research Center, Aalborg University Hospital , Denmark and Department of Clinical Medicine , Aalborg University , Denmark
| |
Collapse
|