1
|
Bile F, Sparaco M, Ruocco E, Miele G, Maida E, Vele R, Mele D, Bonavita S, Lavorgna L. Dermatological Neoplastic Diseases Complicating Treatment with Monoclonal Antibodies for Multiple Sclerosis. J Clin Med 2024; 13:5133. [PMID: 39274345 PMCID: PMC11396336 DOI: 10.3390/jcm13175133] [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: 07/18/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/16/2024] Open
Abstract
Background: Over the past 20 years, the treatment scenario of multiple sclerosis (MS) has radically changed, and an ever-increasing number of disease-modifying treatments has emerged. Among high-efficacy treatment agents, monoclonal antibodies (mAbs) have become a mainstay in a MS patient's treatment due to their targeted mechanism, high efficacy, and favorable risk profile. The latter varies from drug to drug and a skin cancer warning has emerged with sphingosine 1-phosphate receptor inhibitors. Several cases of skin malignancy in people with MS (pwMS) undergoing therapy with mAbs have also been described, but dermatological follow-up is not currently indicated. Objectives: The aim of this review is to investigate cases of cutaneous malignancy during mAb therapy and to explore possible pathophysiological mechanisms to evaluate the potential need for regular dermatological follow-ups in pwMS treated with mAbs. Methods: A literature search for original articles and reviews in PubMed was conducted with no date restrictions. Results: A total of 1019 results were retrieved. Duplicates were removed using Endnote and manually. Only peer-reviewed studies published in English were considered for inclusion. At the end of these screening procedures, 54 studies published between 2001 and 2024 that met the objectives of this review were selected and reported. Conclusions: The available data do not show a clear link between monoclonal antibody (mAb) treatment in pwMS and the risk of skin cancer. At present, these treatments remain contraindicated for people with cancer. Dermatological screening is advisable before starting mAb treatment in pwMS, and subsequent follow-ups should be individualized according to each patient's risk profile.
Collapse
Affiliation(s)
- Floriana Bile
- Department of Advanced Medical and Surgical Sciences, 2nd Division of Neurology, University of Campania Luigi Vanvitelli, Via Sergio Pansini, 5, 80131 Naples, Italy
| | - Maddalena Sparaco
- 2nd Division of Neurology, University Hospital of Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Eleonora Ruocco
- Dermatology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Giuseppina Miele
- 2nd Division of Neurology, University Hospital of Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Elisabetta Maida
- Department of Advanced Medical and Surgical Sciences, 2nd Division of Neurology, University of Campania Luigi Vanvitelli, Via Sergio Pansini, 5, 80131 Naples, Italy
| | - Renato Vele
- Department of Advanced Medical and Surgical Sciences, 2nd Division of Neurology, University of Campania Luigi Vanvitelli, Via Sergio Pansini, 5, 80131 Naples, Italy
| | - Davide Mele
- Department of Advanced Medical and Surgical Sciences, 2nd Division of Neurology, University of Campania Luigi Vanvitelli, Via Sergio Pansini, 5, 80131 Naples, Italy
| | - Simona Bonavita
- Department of Advanced Medical and Surgical Sciences, 2nd Division of Neurology, University of Campania Luigi Vanvitelli, Via Sergio Pansini, 5, 80131 Naples, Italy
| | - Luigi Lavorgna
- 1st Division of Neurology, University Hospital of Campania Luigi Vanvitelli, 80138 Naples, Italy
| |
Collapse
|
2
|
Canto-Gomes J, Boleixa D, Teixeira C, Martins da Silva A, González-Suárez I, Cerqueira J, Correia-Neves M, Nobrega C. Distinct disease-modifying therapies are associated with different blood immune cell profiles in people with relapsing-remitting multiple sclerosis. Int Immunopharmacol 2024; 131:111826. [PMID: 38461632 DOI: 10.1016/j.intimp.2024.111826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 03/12/2024]
Abstract
Disease modifying therapies (DMTs) used for treating people with relapsing-remitting multiple sclerosis (pwRRMS) target the immune system by different mechanisms of action. However, there is a lack of a comprehensive assessment of their effects on the immune system in comparison to treatment-naïve pwRRMS. Herein, we evaluated the numbers of circulating B cells, CD4+ and CD8+ T cells, regulatory T cells (Tregs), natural killer (NK) cells and NKT cells, and their subsets, in pwRRMS who were treatment-naïve or treated with different DMTs. Compared to treatment-naïve pwRRMS, common and divergent effects on immune system cells were observed on pwRRMS treated with different DMTs, with no consistent pattern across all therapies in any of the cell populations analysed. PwRRMS treated with fingolimod, dimethyl fumarate (DMF), or alemtuzumab have reduced numbers of CD4+ and CD8+ T cells, as well as Treg subsets, with fingolimod causing the most pronounced decrease in T cell subsets. In contrast, teriflunomide and interferon (IFN) β have minimal impact on T cells, and natalizumab marginally increases the number of memory T cells in the blood. The effect of DMTs on the B cell, NKT and NK cell subsets is highly variable with alemtuzumab inducing a strong increase in the number of the most immature NK cells and its subsets. This study comprehensively evaluates the magnitude of the effect of different DMTs on blood immune cells providing a better understanding of therapy outcome. Furthermore, the lack of a discernible pattern in the effects of DMTs on blood immune cells suggests that multiple immune cells can independently modulate the disease.
Collapse
Affiliation(s)
- João Canto-Gomes
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's, PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Daniela Boleixa
- Porto University Hospital Center, Porto, Portugal; Multidisciplinary Unit for Biomedical Research (UMIB) - ICBAS, University of Porto, Porto, Portugal
| | - Catarina Teixeira
- Porto University Hospital Center, Porto, Portugal; Multidisciplinary Unit for Biomedical Research (UMIB) - ICBAS, University of Porto, Porto, Portugal
| | - Ana Martins da Silva
- Porto University Hospital Center, Porto, Portugal; Multidisciplinary Unit for Biomedical Research (UMIB) - ICBAS, University of Porto, Porto, Portugal
| | - Inés González-Suárez
- Álvaro Cunqueiro Hospital, Vigo, Spain; University Hospital Complex of Vigo, Vigo, Spain
| | - João Cerqueira
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's, PT Government Associate Laboratory, Braga, Guimarães, Portugal; Hospital of Braga, Braga, Portugal; Clinical Academic Centre, Hospital of Braga, Braga, Portugal
| | - Margarida Correia-Neves
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's, PT Government Associate Laboratory, Braga, Guimarães, Portugal; Division of Infectious Diseases and Center for Molecular Medicine, Department of Medicine Solna, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Claudia Nobrega
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's, PT Government Associate Laboratory, Braga, Guimarães, Portugal.
| |
Collapse
|
3
|
Schmassmann P, Roux J, Dettling S, Hogan S, Shekarian T, Martins TA, Ritz MF, Herter S, Bacac M, Hutter G. Single-cell characterization of human GBM reveals regional differences in tumor-infiltrating leukocyte activation. eLife 2023; 12:RP92678. [PMID: 38127790 PMCID: PMC10735226 DOI: 10.7554/elife.92678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Abstract
Glioblastoma (GBM) harbors a highly immunosuppressive tumor microenvironment (TME) which influences glioma growth. Major efforts have been undertaken to describe the TME on a single-cell level. However, human data on regional differences within the TME remain scarce. Here, we performed high-depth single-cell RNA sequencing (scRNAseq) on paired biopsies from the tumor center, peripheral infiltration zone and blood of five primary GBM patients. Through analysis of >45,000 cells, we revealed a regionally distinct transcription profile of microglia (MG) and monocyte-derived macrophages (MdMs) and an impaired activation signature in the tumor-peripheral cytotoxic-cell compartment. Comparing tumor-infiltrating CD8+ T cells with circulating cells identified CX3CR1high and CX3CR1int CD8+ T cells with effector and memory phenotype, respectively, enriched in blood but absent in the TME. Tumor CD8+ T cells displayed a tissue-resident memory phenotype with dysfunctional features. Our analysis provides a regionally resolved mapping of transcriptional states in GBM-associated leukocytes, serving as an additional asset in the effort towards novel therapeutic strategies to combat this fatal disease.
Collapse
Affiliation(s)
- Philip Schmassmann
- Brain Tumor Immunotherapy Lab, Department of Biomedicine, University of BaselBaselSwitzerland
| | - Julien Roux
- Bioinformatics Core Facility, Department of Biomedicine, University of BaselBaselSwitzerland
- Swiss Institute of BioinformaticsBaselSwitzerland
| | - Steffen Dettling
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center MunichPenzbergGermany
| | - Sabrina Hogan
- Brain Tumor Immunotherapy Lab, Department of Biomedicine, University of BaselBaselSwitzerland
| | - Tala Shekarian
- Brain Tumor Immunotherapy Lab, Department of Biomedicine, University of BaselBaselSwitzerland
| | - Tomás A Martins
- Brain Tumor Immunotherapy Lab, Department of Biomedicine, University of BaselBaselSwitzerland
| | - Marie-Françoise Ritz
- Brain Tumor Immunotherapy Lab, Department of Biomedicine, University of BaselBaselSwitzerland
| | - Sylvia Herter
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center ZürichSchlierenSwitzerland
| | - Marina Bacac
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center ZürichSchlierenSwitzerland
| | - Gregor Hutter
- Brain Tumor Immunotherapy Lab, Department of Biomedicine, University of BaselBaselSwitzerland
- Department of Neurosurgery, University Hospital BaselBaselSwitzerland
| |
Collapse
|
4
|
Zhang Q, Zhang S, Chen J, Xie Z. The Interplay between Integrins and Immune Cells as a Regulator in Cancer Immunology. Int J Mol Sci 2023; 24:6170. [PMID: 37047140 PMCID: PMC10093897 DOI: 10.3390/ijms24076170] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 04/14/2023] Open
Abstract
Integrins are a group of heterodimers consisting of α and β subunits that mediate a variety of physiological activities of immune cells, including cell migration, adhesion, proliferation, survival, and immunotolerance. Multiple types of integrins act differently on the same immune cells, while the same integrin may exert various effects on different immune cells. In the development of cancer, integrins are involved in the regulation of cancer cell proliferation, invasion, migration, and angiogenesis; conversely, integrins promote immune cell aggregation to mediate the elimination of tumors. The important roles of integrins in cancer progression have provided valuable clues for the diagnosis and targeted treatment of cancer. Furthermore, many integrin inhibitors have been investigated in clinical trials to explore effective regimens and reduce side effects. Due to the complexity of the mechanism of integrin-mediated cancer progression, challenges remain in the research and development of cancer immunotherapies (CITs). This review enumerates the effects of integrins on four types of immune cells and the potential mechanisms involved in the progression of cancer, which will provide ideas for more optimal CIT in the future.
Collapse
Affiliation(s)
- Qingfang Zhang
- College of Basic Medical, Nanchang University, Nanchang 330006, China
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Shuo Zhang
- College of Basic Medical, Nanchang University, Nanchang 330006, China
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Jianrui Chen
- College of Basic Medical, Nanchang University, Nanchang 330006, China
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Zhenzhen Xie
- College of Basic Medical, Nanchang University, Nanchang 330006, China
| |
Collapse
|
5
|
Zheleznyak A, Mixdorf M, Marsala L, Prior J, Yang X, Cui G, Xu B, Fletcher S, Fontana F, Lanza G, Achilefu S. Orthogonal targeting of osteoclasts and myeloma cells for radionuclide stimulated dynamic therapy induces multidimensional cell death pathways. Theranostics 2021; 11:7735-7754. [PMID: 34335961 PMCID: PMC8315072 DOI: 10.7150/thno.60757] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 06/09/2021] [Indexed: 12/13/2022] Open
Abstract
Rationale: Multiple myeloma (MM) is a multifocal malignancy of bone marrow plasma cells, characterized by vicious cycles of remission and relapse that eventually culminate in death. The disease remains mostly incurable largely due to the complex interactions between the bone microenvironment (BME) and MM cells (MMC). In the “vicious cycle” of bone disease, abnormal activation of osteoclasts (OCs) by MMC causes severe osteolysis, promotes immune evasion, and stimulates the growth of MMC. Disrupting these cancer-stroma interactions would enhance treatment response. Methods: To disrupt this cycle, we orthogonally targeted nanomicelles (NM) loaded with non-therapeutic doses of a photosensitizer, titanocene (TC), to VLA-4 (α4ß1, CD49d/CD29) expressing MMC (MM1.S) and αvß3 (CD51/CD61) expressing OC. Concurrently, a non-lethal dose of a radiopharmaceutical, 18F-fluorodeoxyglucose ([18F]FDG) administered systemically interacted with TC (radionuclide stimulated therapy, RaST) to generate cytotoxic reactive oxygen species (ROS). The in vitro and in vivo effects of RaST were characterized in MM1.S cell line, as well as in xenograft and isograft MM animal models. Results: Our data revealed that RaST induced non-enzymatic hydroperoxidation of cellular lipids culminating in mitochondrial dysfunction, DNA fragmentation, and caspase-dependent apoptosis of MMC using VLA-4 avid TC-NMs. RaST upregulated the expression of BAX, Bcl-2, and p53, highlighting the induction of apoptosis via the BAK-independent pathway. The enhancement of multicopper oxidase enzyme F5 expression, which inhibits lipid hydroperoxidation and Fenton reaction, was not sufficient to overcome RaST-induced increase in the accumulation of irreversible function-perturbing α,ß-aldehydes that exerted significant and long-lasting damage to both DNA and proteins. In vivo, either VLA-4-TC-NM or αvß3-TC-NMs RaST induced a significant therapeutic effect on immunocompromised but not immunocompetent MM-bearing mouse models. Combined treatment with both VLA-4-TC-NM and αvß3-TC-NMs synergistically inhibited osteolysis, reduced tumor burden, and prevented rapid relapse in both in vivo models of MM. Conclusions: By targeting MM and bone cells simultaneously, combination RaST suppressed MM disease progression through a multi-prong action on the vicious cycle of bone cancer. Instead of using the standard multidrug approach, our work reveals a unique photophysical treatment paradigm that uses nontoxic doses of a single light-sensitive drug directed orthogonally to cancer and bone cells, followed by radionuclide-stimulated generation of ROS to inhibit tumor progression and minimize osteolysis in both immunocompetent murine and immunocompromised human MM models.
Collapse
|
6
|
Carbone ML, Lacal PM, Messinese S, De Giglio L, Pozzilli C, Persechino S, Mazzanti C, Failla CM, Pagnanelli G. Multiple Sclerosis Treatment and Melanoma Development. Int J Mol Sci 2020; 21:E2950. [PMID: 32331328 PMCID: PMC7216218 DOI: 10.3390/ijms21082950] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/19/2020] [Accepted: 04/20/2020] [Indexed: 11/16/2022] Open
Abstract
Therapy of multiple sclerosis (MS) with disease-modifying agents such as natalizumab or fingolimod has been associated with the development of cutaneous melanoma. Here we briefly revise literature data and report of a case of a 48-year old woman who developed a melanoma and several atypical naevi after sub sequential treatment with natalizumab (1 year) and fingolimod (7 years). By immunohistochemistry we observed the presence of T cells and leukocyte infiltration as well as of vascular endothelial growth factor (VEGF)-A expression in the patient melanoma biopsy. Then, we analyzed proliferation, migration and VEGF-A expression in three melanoma cell lines and found out that both natalizumab and fingolimod inhibited tumor cell proliferation but promoted or blocked cell migration depending on the cell line examined. VEGF-A secretion was augmented in one melanoma cell line only after fingolimod treatment. In conclusion, our in vitro data do not support the hypothesis of a direct action of natalizumab or fingolimod on melanoma progression but acting on the tumor microenvironment these treatments could indirectly favor melanoma evolution.
Collapse
Affiliation(s)
| | | | - Serena Messinese
- I Dermatology Department, IDI-IRCCS, 00167 Rome, Italy; (S.M.); (C.M.); (G.P.)
| | - Laura De Giglio
- Medicine Department, Neurology Unit, San Filippo Neri Hospital, 00135 Rome, Italy;
| | - Carlo Pozzilli
- Department of Human Neurosciences, Sant’Andrea Hospital, MS Centre, Sapienza University, 00189 Rome, Italy;
| | - Severino Persechino
- NESMOS Department, Dermatology Unit, Sant’Andrea Hospital, Sapienza University, 00189 Rome, Italy;
| | - Cinzia Mazzanti
- I Dermatology Department, IDI-IRCCS, 00167 Rome, Italy; (S.M.); (C.M.); (G.P.)
| | | | - Gianluca Pagnanelli
- I Dermatology Department, IDI-IRCCS, 00167 Rome, Italy; (S.M.); (C.M.); (G.P.)
| |
Collapse
|
7
|
Amoriello R, Greiff V, Aldinucci A, Bonechi E, Carnasciali A, Peruzzi B, Repice AM, Mariottini A, Saccardi R, Mazzanti B, Massacesi L, Ballerini C. The TCR Repertoire Reconstitution in Multiple Sclerosis: Comparing One-Shot and Continuous Immunosuppressive Therapies. Front Immunol 2020; 11:559. [PMID: 32328061 PMCID: PMC7160336 DOI: 10.3389/fimmu.2020.00559] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 03/11/2020] [Indexed: 11/13/2022] Open
Abstract
Natalizumab (NTZ) and autologous hematopoietic stem cell transplantation (AHSCT) are two successful treatments for relapsing-remitting multiple sclerosis (RRMS), an autoimmune T-cell-driven disorder affecting the central nervous system that is characterized by relapses interspersed with periods of complete or partial recovery. Both RRMS treatments have been documented to impact T-cell subpopulations and the T-cell receptor (TCR) repertoire in terms of clone frequency, but, so far, the link between T-cell naive and memory populations, autoimmunity, and treatment outcome has not yet been established hindering insight into the post-treatment TCR landscape of MS patients. To address this important knowledge gap, we tracked peripheral T-cell subpopulations (naïve and memory CD4+ and CD8+) across 15 RRMS patients before and after two years of continuous treatment (NTZ) and a single treatment course (AHSCT) by high-throughput TCRß sequencing. We found that the two MS treatments left treatment-specific multidimensional traces in patient TCRß repertoire dynamics with respect to clonal expansion, clonal diversity and repertoire architecture. Comparing MS TCR sequences with published datasets suggested that the majority of public TCRs belonged to virus-associated sequences. In summary, applying multi-dimensional computational immunology to a TCRß dataset of treated MS patients, we show that qualitative changes of TCRß repertoires encode treatment-specific information that may be relevant for future clinical trials monitoring and personalized MS follow-up, diagnosis and treatment regimes. Natalizumab (NTZ) and autologous hematopoietic stem cell transplantation (AHSCT) are two successful treatments for relapsing-remitting multiple sclerosis (RRMS), an autoimmune T-cell-driven disorder affecting the central nervous system that is characterized by relapses interspersed with periods of complete or partial recovery. Both RRMS treatments have been documented to impact T-cell subpopulations and the T-cell receptor (TCR) repertoire in terms of clone frequency, but, so far, the link between T-cell naive and memory populations, autoimmunity, and treatment outcome has not yet been established hindering insight into the posttreatment TCR landscape of MS patients. To address this important knowledge gap, we tracked peripheral T-cell subpopulations (naive and memory CD4+ and CD8+) across 15 RRMS patients before and after 2 years of continuous treatment (NTZ) and a single treatment course (AHSCT) by high-throughput TCRβ sequencing. We found that the two MS treatments left treatment-specific multidimensional traces in patient TCRβ repertoire dynamics with respect to clonal expansion, clonal diversity, and repertoire architecture. Comparing MS TCR sequences with published datasets suggested that the majority of public TCRs belonged to virus-associated sequences. In summary, applying multidimensional computational immunology to a TCRβ dataset of treated MS patients, we show that qualitative changes of TCRβ repertoires encode treatment-specific information that may be relevant for future clinical trials monitoring and personalized MS follow-up, diagnosis, and treatment regimens.
Collapse
Affiliation(s)
- Roberta Amoriello
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino (NEUROFARBA), University of Florence, Florence, Italy
| | - Victor Greiff
- Department of Immunology, University of Oslo, Oslo, Norway
| | - Alessandra Aldinucci
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino (NEUROFARBA), University of Florence, Florence, Italy
| | - Elena Bonechi
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino (NEUROFARBA), University of Florence, Florence, Italy
| | - Alberto Carnasciali
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino (NEUROFARBA), University of Florence, Florence, Italy
| | - Benedetta Peruzzi
- Centro Diagnostico di Citofluorimetria e Immunoterapia, Careggi University Hospital, Florence, Italy
| | - Anna Maria Repice
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino (NEUROFARBA), University of Florence, Florence, Italy
| | - Alice Mariottini
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino (NEUROFARBA), University of Florence, Florence, Italy
| | - Riccardo Saccardi
- SODc Terapie Cellulari e Medicina Trasfusionale, Careggi University Hospital, Florence, Italy
| | - Benedetta Mazzanti
- Dipartimento di Medicina Sperimentale e Clinica (DMSC), University of Florence, Florence, Italy
| | - Luca Massacesi
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino (NEUROFARBA), University of Florence, Florence, Italy
| | - Clara Ballerini
- Dipartimento di Medicina Sperimentale e Clinica (DMSC), University of Florence, Florence, Italy
| |
Collapse
|
8
|
Adverse events and monitoring requirements associated with monoclonal antibody therapy in patients with multiple sclerosis. DRUGS & THERAPY PERSPECTIVES 2019. [DOI: 10.1007/s40267-019-00682-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
9
|
Sotgiu S, Eusebi A, Begliuomini C, Guerini FR, Carta A. Reader response: Pregnancy decision-making in women with multiple sclerosis treated with natalizumab: I: Fetal risks. Neurology 2019; 91:850. [PMID: 30373925 DOI: 10.1212/wnl.0000000000006429] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
10
|
Chen YL, Qiao YC, Xu Y, Ling W, Pan YH, Huang YC, Geng LJ, Zhao HL, Zhang XX. Serum TNF-α concentrations in type 2 diabetes mellitus patients and diabetic nephropathy patients: A systematic review and meta-analysis. Immunol Lett 2017; 186:52-58. [PMID: 28414180 DOI: 10.1016/j.imlet.2017.04.003] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 03/25/2017] [Accepted: 04/10/2017] [Indexed: 11/24/2022]
Abstract
OBJECTIVES The aim of this study was to investigate whether the concentrations of serum tumor necrosis factor-α (TNF-α), a pro-inflammatory cytokine, increased in type 2 diabetes mellitus (T2DM) and type 2 diabetic nephropathy (T2DN) patients. METHODS The four databases (PubMed, CNKI, WanFang and Chinese-Cqvip) were searched from Jan 1, 1999 to October 1, 2016 for all clinical case-control studies about the serum TNF-α concentrations in T2DM and T2DN patients. All relevant data were extracted from published reports. The meta-analysis was performed to compare the changes of serum TNF-α concentrations of T2DN and T2DM patients in Eastern and Western with healthy controls. We further evaluated concentrations of serum TNF-α in T2DN patients with mincroalbuminuria or macroalbuminuria. Random-effects models were adopted to assess the pooling data among various variations. RESULTS In total of 6 studies (744 patients and 277 healthy controls) were included in this study. Compared with healthy controls (both p<0.01), the groups of different albuminuria levels and ethnicities both showed that the serum TNF-α levels were significantly elevated in T2DN patients as well as in eastern T2DN patients (p=0.001), but not significant changed in western T2DN patients (p=0.081). The results were stable through sensitivity analysis and no significant publications bias existed in this meta-analysis. CONCLUSIONS Serum TNF-α concentrations are obviously increased in T2DN and T2DM patients, but higher in T2DN patients, suggesting an elevated inflammatory burden in T2DN patients.
Collapse
Affiliation(s)
- Yin-Ling Chen
- Center of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China; Department of Immunology, Faculty of Basic Medicine, Guilin Medical University, Guilin 541004, China
| | - Yong-Chao Qiao
- Department of Immunology, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Yan Xu
- Department of Immunology, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Wei Ling
- Center of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China
| | - Yan-Hong Pan
- Center of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China; Department of Immunology, Faculty of Basic Medicine, Guilin Medical University, Guilin 541004, China
| | - Yong-Cheng Huang
- Center of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China
| | - Li-Jun Geng
- Center of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China; Department of Immunology, Faculty of Basic Medicine, Guilin Medical University, Guilin 541004, China
| | - Hai-Lu Zhao
- Center of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China; Department of Immunology, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China; Department of Immunology, Faculty of Basic Medicine, Guilin Medical University, Guilin 541004, China
| | - Xiao-Xi Zhang
- Center of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China; Department of Immunology, Faculty of Basic Medicine, Guilin Medical University, Guilin 541004, China.
| |
Collapse
|