Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Mult Scler Relat Disord ; 82: 105400, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38181696

RESUMO

Neutropenia serves as a risk factor for severe infection and is a consequence of some immune-depleting immunotherapies. This occurs in people with multiple sclerosis following chemotherapy-conditioning in haematopoietic stem cell transplantation and potent B cell targeting agents. Whilst CD52 is expressed by neutrophils and may contribute to early-onset neutropenia following alemtuzumab treatment, deoxycytidine kinase and CD20 antigen required for activity of cladribine tablets, off-label rituximab, ocrelizumab, ofatumumab and ublituximab are not or only weakly expressed by neutrophils. Therefore, alternative explanations are needed for the rare occurrence of early and late-onset neutropenia following such treatments. This probably occurs due to alterations in the balance of granulopoiesis and neutrophil removal. Neutrophils are short-lived, and their removal may be influenced by drug-associated infections, the killing mechanisms of the therapies and amplified by immune dyscrasia due to influences on neutropoiesis following growth factor rerouting for B cell recovery and cytokine deficits following lymphocyte depletion. This highlights the small but evident neutropenia risks following sustained B cell depletion with some treatments.


Assuntos
Esclerose Múltipla , Neutropenia , Humanos , Esclerose Múltipla/terapia , Alemtuzumab/efeitos adversos , Rituximab/efeitos adversos , Fatores Imunológicos/efeitos adversos , Neutropenia/induzido quimicamente , Antígenos CD20
2.
Mult Scler ; 29(8): 979-989, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37431627

RESUMO

BACKGROUND: People with multiple sclerosis (pwMS) treated with certain disease-modifying therapies (DMTs) have attenuated IgG response following COVID-19 vaccination; however, the clinical consequences remain unclear. OBJECTIVE: To report COVID-19 rates in pwMS according to vaccine serology. METHODS: PwMS with available (1) serology 2-12 weeks following COVID-19 vaccine 2 and/or vaccine 3 and (2) clinical data on COVID-19 infection/hospitalisation were included. Logistic regression was performed to examine whether seroconversion following vaccination predicted risk of subsequent COVID-19 infection after adjusting for potential confounders. Rates of severe COVID-19 (requiring hospitalisation) were also calculated. RESULTS: A total of 647 pwMS were included (mean age 48 years, 500 (77%) female, median Expanded Disability Status Scale (EDSS) 3.5% and 524 (81%) exposed to DMT at the time of vaccine 1). Overall, 472 out of 588 (73%) were seropositive after vaccines 1 and 2 and 222 out of 305 (73%) after vaccine 3. Seronegative status after vaccine 2 was associated with significantly higher odds of subsequent COVID-19 infection (odds ratio (OR): 2.35, 95% confidence interval (CI): 1.34-4.12, p = 0.0029), whereas seronegative status after vaccine 3 was not (OR: 1.05, 95% CI: 0.57-1.91). Five people (0.8%) experienced severe COVID-19, all of whom were seronegative after most recent vaccination. CONCLUSION: Attenuated humoral response to initial COVID-19 vaccination predicts increased risk of COVID-19 in pwMS, but overall low rates of severe COVID-19 were seen.


Assuntos
Vacinas contra COVID-19 , COVID-19 , Esclerose Múltipla , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , COVID-19/epidemiologia , COVID-19/prevenção & controle , Vacinas contra COVID-19/efeitos adversos , Hospitalização , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/epidemiologia , Vacinação
3.
J Mol Biol ; 435(15): 168146, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37201677

RESUMO

One infection method widely used by many gram-negative bacteria involves a protein nanomachine called the Type Three Secretion System (T3SS). The T3SS enables the transportation of bacterial "toxins" via a proteinaceous channel that directly links the cytosol of the bacteria and host cell. The channel from the bacteria is completed by a translocon pore formed by two proteins named the major and minor translocators. Prior to pore formation, the translocator proteins are bound to a small chaperone within the bacterial cytoplasm. This interaction is crucial to effective secretion. Here we investigated the specificity of the binding interfaces of the translocator-chaperone complexes from Pseudomonas aeruginosa via the selection of peptide and protein libraries based on its chaperone PcrH. Five libraries encompassing PcrH's N-terminal and central α-helices were panned, using ribosome display, against both the major (PopB) and minor (PopD) translocator. Both translocators were shown to significantly enrich a similar pattern of WT and non-WT sequences from the libraries. This highlighted key similarities/differences between the interactions of the major and minor translocators with their chaperone. Moreover, as the enriched non-WT sequences were specific to each translocator, it would suggest that PcrH can be adapted to bind each translocator individually. The ability to evolve such proteins indicates that these molecules may provide promising anti-bacterial candidates.


Assuntos
Proteínas de Bactérias , Chaperonas Moleculares , Pseudomonas aeruginosa , Sistemas de Secreção Tipo III , Braço , Proteínas de Bactérias/química , Toxinas Bacterianas/metabolismo , Citoplasma/metabolismo , Chaperonas Moleculares/química , Ligação Proteica , Pseudomonas aeruginosa/metabolismo , Sistemas de Secreção Tipo III/química
4.
Biochemistry ; 62(8): 1420-1427, 2023 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-36996474

RESUMO

The type III secretion system is a large multiprotein complex that many Gram-negative bacteria use for infection. A crucial part of the complex is its translocon pore formed by two proteins: the major and minor translocators. The pore completes a proteinaceous channel from the bacterial cytosol through the host cell membrane and allows the direct injection of bacterial toxins. Effective pore formation is predicated by the translocator proteins binding to a small chaperone within the bacterial cytoplasm. Given the vital role of the chaperone-translocator interaction, we investigated the specificity of the "N-terminal anchor" binding interface present in both translocator-chaperone complexes from Pseudomonas aeruginosa. Isothermal calorimetry (ITC), alanine scanning, and the selection of a motif-based peptide library using ribosome display were used to characterize the major (PopB) and minor (PopD) translocator interactions with their chaperone PcrH. We show that 10 mer PopB51-60 and PopD47-56 peptides bind to PcrH with a KD of 148 ± 18 and 91 ± 9 µM, respectively. Moreover, mutation to alanine of each of the consensus residues (xxVxLxxPxx) of the PopB peptide severely affected or completely abrogated binding to PcrH. When the directed peptide library (X-X-hydrophobic-X-L-X-X-P-X-X) was panned against PcrH, there was no obvious convergence at the varied residues. The PopB/PopD wild-type (WT) sequences were also not prevalent. However, a consensus peptide was shown to bind to PcrH with micromolar affinity. Thus, selected sequences were binding with similar affinities to WT PopB/PopD peptides. These results demonstrate that only the conserved "xxLxxP" motif drives binding at this interface.


Assuntos
Proteínas de Bactérias , Pseudomonas aeruginosa , Pseudomonas aeruginosa/metabolismo , Proteínas de Bactérias/química , Biblioteca de Peptídeos , Chaperonas Moleculares/metabolismo , Citoplasma/metabolismo , Peptídeos/metabolismo
5.
Mult Scler Relat Disord ; 69: 104425, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36470168

RESUMO

BACKGROUND: Sphingosine-one phosphate receptor (S1PR) modulation inhibits S1PR1-mediated lymphocyte migration, lesion formation and positively-impacts on active multiple sclerosis (MS). These S1PR modulatory drugs have different: European Union use restrictions, pharmacokinetics, metabolic profiles and S1PR receptor affinities that may impact MS-management. Importantly, these confer useful properties in dealing with COVID-19, anti-viral drug responses and generating SARS-CoV-2 vaccine responses. OBJECTIVE: To examine the biology and emerging data that potentially underpins immunity to the SARS-CoV-2 virus following natural infection and vaccination and determine how this impinges on the use of current sphingosine-one-phosphate modulators used in the treatment of MS. METHODS: A literature review was performed, and data on infection, vaccination responses; S1PR distribution and functional activity was extracted from regulatory and academic information within the public domain. OBSERVATIONS: Most COVID-19 related information relates to the use of fingolimod. This indicates that continuous S1PR1, S1PR3, S1PR4 and S1PR5 modulation is not associated with a worse prognosis following SARS-CoV-2 infection. Whilst fingolimod use is associated with blunted seroconversion and reduced peripheral T-cell vaccine responses, it appears that people on siponimod, ozanimod and ponesimod exhibit stronger vaccine-responses, which could be related notably to a limited impact on S1PR4 activity. Whilst it is thought that S1PR3 controls B cell function in addition to actions by S1PR1 and S1PR2, this may be species-related effect in rodents that is not yet substantiated in humans, as seen with bradycardia issues. Blunted antibody responses can be related to actions on B and T-cell subsets, germinal centre function and innate-immune biology. Although S1P1R-related functions are seeming central to control of MS and the generation of a fully functional vaccination response; the relative lack of influence on S1PR4-mediated actions on dendritic cells may increase the rate of vaccine-induced seroconversion with the newer generation of S1PR modulators and improve the risk-benefit balance IMPLICATIONS: Although fingolimod is a useful asset in controlling MS, recently-approved S1PR modulators may have beneficial biology related to pharmacokinetics, metabolism and more-restricted targeting that make it easier to generate infection-control and effective anti-viral responses to SARS-COV-2 and other pathogens. Further studies are warranted.


Assuntos
COVID-19 , Esclerose Múltipla , Moduladores do Receptor de Esfingosina 1 Fosfato , Humanos , Cloridrato de Fingolimode/farmacologia , Cloridrato de Fingolimode/uso terapêutico , Moduladores do Receptor de Esfingosina 1 Fosfato/uso terapêutico , COVID-19/prevenção & controle , Vacinas contra COVID-19 , SARS-CoV-2 , Receptores de Esfingosina-1-Fosfato/uso terapêutico , Esfingosina , Vacinação
6.
Mult Scler Relat Disord ; 64: 103937, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35700625

RESUMO

BACKGROUND: People with MS treated with anti-CD20 therapies and fingolimod often have attenuated responses to initial COVID-19 vaccination. However, uncertainties remain about the benefit of a 3rd (booster) COVID-19 vaccine in this group. METHODS: PwMS without a detectable IgG response following COVID-19 vaccines 1&2 were invited to participate. Participants provided a dried blood spot +/- venous blood sample 2-12 weeks following COVID-19 vaccine 3. Humoral and T cell responses to SARS-CoV-2 spike protein and nucleocapsid antigen were measured. RESULTS: Of 81 participants, 79 provided a dried blood spot sample, of whom 38 also provided a whole blood sample; 2 provided only whole blood. Anti-SARS-CoV-2-spike IgG seroconversion post-COVID-19 vaccine 3 occurred in 26/79 (33%) participants; 26/40 (65%) had positive T-cell responses. Overall, 31/40 (78%) demonstrated either humoral or cellular immune response post-COVID-19 vaccine 3. There was no association between laboratory evidence of prior COVID-19 and seroconversion following vaccine 3. CONCLUSIONS: Approximately one third of pwMS who were seronegative after initial COVID-19 vaccination seroconverted after booster (third) vaccination, supporting the use of boosters in this group. Almost 8 out of 10 had a measurable immune response following 3rd COVID-19 vaccine.


Assuntos
COVID-19 , Esclerose Múltipla , Anticorpos Antivirais , COVID-19/prevenção & controle , Vacinas contra COVID-19 , Humanos , Imunoglobulina G , Esclerose Múltipla/tratamento farmacológico , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Vacinação
7.
Clin Exp Immunol ; 207(3): 263-271, 2022 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-35553629

RESUMO

Although there is an ever-increasing number of disease-modifying treatments for relapsing multiple sclerosis (MS), few appear to influence coronavirus disease 2019 (COVID-19) severity. There is concern about the use of anti-CD20-depleting monoclonal antibodies, due to the apparent increased risk of severe disease following severe acute respiratory syndrome corona virus two (SARS-CoV-2) infection and inhibition of protective anti-COVID-19 vaccine responses. These antibodies are given as maintenance infusions/injections and cause persistent depletion of CD20+ B cells, notably memory B-cell populations that may be instrumental in the control of relapsing MS. However, they also continuously deplete immature and mature/naïve B cells that form the precursors for infection-protective antibody responses, thus blunting vaccine responses. Seroconversion and maintained SARS-CoV-2 neutralizing antibody levels provide protection from COVID-19. However, it is evident that poor seroconversion occurs in the majority of individuals following initial and booster COVID-19 vaccinations, based on standard 6 monthly dosing intervals. Seroconversion may be optimized in the anti-CD20-treated population by vaccinating prior to treatment onset or using extended/delayed interval dosing (3-6 month extension to dosing interval) in those established on therapy, with B-cell monitoring until (1-3%) B-cell repopulation occurs prior to vaccination. Some people will take more than a year to replete and therefore protection may depend on either the vaccine-induced T-cell responses that typically occur or may require prophylactic, or rapid post-infection therapeutic, antibody or small-molecule antiviral treatment to optimize protection against COVID-19. Further studies are warranted to demonstrate the safety and efficacy of such approaches and whether or not immunity wanes prematurely as has been observed in the other populations.


Assuntos
COVID-19 , Esclerose Múltipla , Anticorpos Antivirais , Antígenos CD20 , COVID-19/prevenção & controle , Vacinas contra COVID-19 , Humanos , Esclerose Múltipla/tratamento farmacológico , SARS-CoV-2 , Soroconversão , Vacinação
8.
Ann Neurol ; 91(1): 89-100, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34687063

RESUMO

OBJECTIVE: The purpose of this study was to investigate the effect of disease modifying therapies on immune response to severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) vaccines in people with multiple sclerosis (MS). METHODS: Four hundred seventy-three people with MS provided one or more dried blood spot samples. Information about coronavirus disease 2019 (COVID-19) and vaccine history, medical, and drug history were extracted from questionnaires and medical records. Dried blood spots were eluted and tested for antibodies to SARS-CoV-2. Antibody titers were partitioned into tertiles with people on no disease modifying therapy as a reference. We calculated the odds ratio of seroconversion (univariate logistic regression) and compared quantitative vaccine response (Kruskal Wallis) following the SARS-CoV-2 vaccine according to disease modifying therapy. We used regression modeling to explore the effect of vaccine timing, treatment duration, age, vaccine type, and lymphocyte count on vaccine response. RESULTS: Compared to no disease modifying therapy, the use of anti-CD20 monoclonal antibodies (odds ratio = 0.03, 95% confidence interval [CI] = 0.01-0.06, p < 0.001) and fingolimod (odds ratio = 0.04; 95% CI = 0.01-0.12) were associated with lower seroconversion following the SARS-CoV-2 vaccine. All other drugs did not differ significantly from the untreated cohort. Both time since last anti-CD20 treatment and total time on treatment were significantly associated with the response to the vaccination. The vaccine type significantly predicted seroconversion, but not in those on anti-CD20 medications. Preliminary data on cellular T-cell immunity showed 40% of seronegative subjects had measurable anti-SARS-CoV-2 T cell responses. INTERPRETATION: Some disease modifying therapies convey risk of attenuated serological response to SARS-CoV-2 vaccination in people with MS. We provide recommendations for the practical management of this patient group. ANN NEUROL 20219999:n/a-n/a.


Assuntos
Antirreumáticos/uso terapêutico , Vacinas contra COVID-19/imunologia , COVID-19/prevenção & controle , Hospedeiro Imunocomprometido , Esclerose Múltipla/imunologia , Soroconversão/efeitos dos fármacos , Adulto , Anticorpos Antivirais/sangue , Anticorpos Antivirais/efeitos dos fármacos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Esclerose Múltipla/tratamento farmacológico , SARS-CoV-2 , Reino Unido
9.
Mult Scler Relat Disord ; 57: 103448, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34902760

RESUMO

BACKGROUND: Ocrelizumab maintains B-cell depletion via six-monthly dosing. Whilst this controls relapsing multiple sclerosis, it also inhibits seroconversion following SARS-CoV-2 vaccination unlike that seen following alemtuzumab and cladribine treatment. Emerging reports suggest that 1-3% B-cell repopulation facilitates seroconversion after CD20-depletion. OBJECTIVE: To determine the frequency of B-cell repopulation levels during and after ocrelizumab treatment. METHODS: Relapse data, lymphocyte and CD19 B-cell numbers were obtained following requests to clinical trial data-repositories. Information was extracted from the phase II ocrelizumab extension (NCT00676715) trial and the phase III cladribine tablet (NCT00213135) and alemtuzumab (NCT00530348/NCT00548405) trials obtained clinical trial data requests RESULTS: Only 3-5% of people with MS exhibit 1% B-cells at 6 months after the last infusion following 3-4 cycles of ocrelizumab, compared to 50-55% at 9 months, and 85-90% at 12 months. During this time relapses occurred at consistent disease-breakthrough rates compared to people during standard therapy. In contrast most people (90-100%) exhibited more than 1% B-cells during treatment with either cladribine or alemtuzumab. CONCLUSIONS: Most people demonstrate B cell repletion within 3 months of the last treatment of alemtuzumab and cladribine. However, few people repopulate peripheral B-cells with standard ocrelizumab dosing. Controlled studies are warranted to examine a view that delaying the dosing interval by 3-6 months may allow more people to potentially seroconvert after vaccination.


Assuntos
COVID-19 , Esclerose Múltipla Recidivante-Remitente , Esclerose Múltipla , Alemtuzumab/uso terapêutico , Anticorpos Monoclonais Humanizados , Vacinas contra COVID-19 , Cladribina , Humanos , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , SARS-CoV-2 , Vacinação
10.
Hum Antibodies ; 29(4): 255-262, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34397407

RESUMO

Multiple sclerosis is the major demyelinating autoimmune disease of the central nervous system. Relapsing MS can be treated by a number of approved monoclonal antibodies that currently target: CD20, CD25 (withdrawn), CD49d and CD52. These all target potentially pathogenic memory B cell subsets and perhaps functionally inhibit pathogenic T cell function. These consist of chimeric, humanized and fully human antibodies. However, despite humanization it is evident that all of these monoclonal antibodies can induce binding and neutralizing antibodies ranging from < 1% to over 80% within a year of treatment. Importantly, it is evident that monitoring these allow prediction of future treatment-failure in some individuals and treatment cessation and switching therefore potentially limiting disease breakthrough and disability accumulation. In response to the COVID-19 pandemic and the need to avoid hospitals, shortened infusion times and extended dose intervals have been implemented, importantly, subcutaneous delivery of alternative treatments or formulations have been developed to allow for home treatment. Therefore, hospital-based and remote monitoring of ADA could therefore be advantageous to optimize patient responses in the future.


Assuntos
COVID-19 , Esclerose Múltipla , Anticorpos Monoclonais Humanizados , Humanos , Células B de Memória , Esclerose Múltipla/tratamento farmacológico , Pandemias , SARS-CoV-2
11.
Hum Antibodies ; 29(3): 171-178, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34151782

RESUMO

The number of biologic drugs available for the treatment of psoriasis continue to expand. However, being biological proteins and thus potentially immunogenic, there is evidence that anti-drug-antibodies develop against the various therapeutic proteins currently being utilised. Although chimeric antibodies that contain elements of the parental rodent monoclonal antibodies are immunogenic, anti-drug antibodies occur even if the biologic is a fully human protein and these can impact on clinical efficacy and safety. However, there is a wide variation in the reported level of anti-drug-antibodies for the same and different treatments that is highlighting issues with various assays used in anti-drug antibody detection. Here we review the available data on the occurrence of anti-drug antibodies in people with psoriasis treated with biologic agents.


Assuntos
Psoríase , Anticorpos Monoclonais/uso terapêutico , Formação de Anticorpos , Produtos Biológicos/uso terapêutico , Humanos , Psoríase/tratamento farmacológico , Resultado do Tratamento
12.
Hum Antibodies ; 29(4): 225-235, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34151784

RESUMO

PURPOSE OF THE REVIEW: Here we critically evaluate the literature on immunotherapy failure in inflammatory bowel disease patients. In particular anti-drug antibody production, and subsequently loss of response as the primary cause of immunotherapy failure in IBD patients. The benefits of shifting from the "standard" empirical dose escalation approach to therapeutic drug monitoring with anti-TNFα therapy is explored. RECENT FINDINGS: The American Gastroenterology Association and British Society of Gastroenterology both currently recommend the use of reactive therapeutic drug monitoring to guide treatment, following loss of response in inflammatory bowel disease patients with active disease. However, further research is required to prove the efficacy of a proactive therapeutic drug monitoring approach alone in remitted IBD patients. SUMMARY: A combination of personalised monitoring approach for anti-drug antibodies and therapeutic drug monitoring could provide beneficial treatment outcome for people with inflammatory bowel disease by predicting drug failure prior to clinical symptoms and allowing timely switching to an alternative drug.


Assuntos
Produtos Biológicos , Doenças Inflamatórias Intestinais , Produtos Biológicos/uso terapêutico , Monitoramento de Medicamentos , Fármacos Gastrointestinais/uso terapêutico , Humanos , Doenças Inflamatórias Intestinais/tratamento farmacológico , Fator de Necrose Tumoral alfa , Estados Unidos
14.
Artigo em Inglês | MEDLINE | ID: mdl-32499328

RESUMO

OBJECTIVE: To test the hypothesis that antidrug antibodies (ADAs) against alemtuzumab could become relevant after repeated treatments for some individuals, possibly explaining occasional treatment resistance. METHODS: Recombinant alemtuzumab single-chain variable fragment antibody with a dual tandem nanoluciferase reporter linker was made and used to detect binding ADAs. Alemtuzumab immunoglobulin G Alexa Fluor 488 conjugate was used in a competitive binding cell-based assay to detect neutralizing ADAs. The assays were used to retrospectively screen, blinded, banked serum samples from people with MS (n = 32) who had received 3 or more cycles of alemtuzumab. Lymphocyte depletion was measured between baseline and about 1 month postinfusion. RESULTS: The number of individuals showing limited depletion of lymphocytes increased with the number of treatment cycles. Lack of depletion was also a poor prognostic feature for future disease activity. ADA responses were detected in 29/32 (90.6%) individuals. Neutralizing antibodies occurred before the development of limited depletion in 6/7 individuals (18.8% of the whole sample). Preinfusion, ADA levels predicted limited, postinfusion lymphocyte depletion. CONCLUSIONS: Although ADAs to alemtuzumab have been portrayed as being of no clinical significance, alemtuzumab-specific antibodies appear to be clinically relevant for some individuals, although causation remains to be established. Monitoring of lymphocyte depletion and the antidrug response may be of practical value in patients requiring additional cycles of alemtuzumab. ADA detection may help to inform on retreatment or switching to another treatment.


Assuntos
Alemtuzumab/efeitos adversos , Alemtuzumab/imunologia , Anticorpos/sangue , Fatores Imunológicos/efeitos adversos , Fatores Imunológicos/imunologia , Esclerose Múltipla/sangue , Esclerose Múltipla/tratamento farmacológico , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Avaliação de Resultados em Cuidados de Saúde , Estudos Retrospectivos
15.
Mult Scler Relat Disord ; 43: 102174, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32464584

RESUMO

BACKGROUND: SARS-CoV-2 viral infection causes COVID-19 that can result in severe acute respiratory distress syndrome (ARDS), which can cause significant mortality, leading to concern that immunosuppressive treatments for multiple sclerosis and other disorders have significant risks for both infection and ARDS. OBJECTIVE: To examine the biology that potentially underpins immunity to the SARS-Cov-2 virus and the immunity-induced pathology related to COVID-19 and determine how this impinges on the use of current disease modifying treatments in multiple sclerosis. OBSERVATIONS: Although information about the mechanisms of immunity are scant, it appears that monocyte/macrophages and then CD8 T cells are important in eliminating the SARS-CoV-2 virus. This may be facilitated via anti-viral antibody responses that may prevent re-infection. However, viral escape and infection of leucocytes to promote lymphopenia, apparent CD8 T cell exhaustion coupled with a cytokine storm and vascular pathology appears to contribute to the damage in ARDS. IMPLICATIONS: In contrast to ablative haematopoietic stem cell therapy, most multiple-sclerosis-related disease modifying therapies do not particularly target the innate immune system and few have any major long-term impact on CD8 T cells to limit protection against COVID-19. In addition, few block the formation of immature B cells within lymphoid tissue that will provide antibody-mediated protection from (re)infection. However, adjustments to dosing schedules may help de-risk the chance of infection further and reduce the concerns of people with MS being treated during the COVID-19 pandemic.


Assuntos
Betacoronavirus/imunologia , Linfócitos T CD8-Positivos/imunologia , Infecções por Coronavirus/imunologia , Síndrome da Liberação de Citocina/imunologia , Imunossupressores/uso terapêutico , Linfopenia/imunologia , Esclerose Múltipla/terapia , Pneumonia Viral/imunologia , Alemtuzumab/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , Anticorpos Antivirais/imunologia , COVID-19 , Cladribina/uso terapêutico , Crotonatos/uso terapêutico , Desprescrições , Fumarato de Dimetilo/uso terapêutico , Cloridrato de Fingolimode/uso terapêutico , Acetato de Glatiramer/uso terapêutico , Transplante de Células-Tronco Hematopoéticas/métodos , Humanos , Hidroxibutiratos , Evasão da Resposta Imune/imunologia , Imunidade Inata/imunologia , Fatores Imunológicos/uso terapêutico , Interferon beta/uso terapêutico , Macrófagos/imunologia , Monócitos/imunologia , Natalizumab/uso terapêutico , Nitrilas , Pandemias , Síndrome do Desconforto Respiratório/imunologia , SARS-CoV-2 , Toluidinas/uso terapêutico
17.
Front Immunol ; 11: 124, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32117274

RESUMO

Alemtuzumab was designed to reduce the immunogenicity of the parent CD52-specific rat immunoglobulin. Although originally marketed for use in cancer (Mabcampath®), alemtuzumab is currently licensed and formulated for the treatment of relapsing multiple sclerosis (Lemtrada®). Perhaps due to its history as the first humanized antibody, the potential of immunogenicity of the molecule has been considered inconsequential, and anti-drug antibodies (ADA) responses were similarly reported as being clinically insignificant. Nonetheless, despite humanization and depletion of peripheral T and B cells, alemtuzumab probably generates the highest frequency of binding and neutralizing ADA of all humanized antibodies currently in clinical use, and they occur rapidly in a large majority of people with MS (pwMS) on alemtuzumab treatment. These ADA appear to be an inherent issue of the biology of the molecule-and more importantly, the target-such that avoidance of immunogenicity-related effects has been facilitated by the dosing schedule used in clinical practice. At the population level this enables the drug to work in most pwMS, but in some individuals, as we show here, antibody neutralization appears to be sufficiently severe to reduce efficacy and allow disease breakthrough. It is therefore imperative that efficacy of lymphocyte depletion and the anti-drug response is monitored in people requiring additional cycles of treatment, notably following disease breakthrough. This may help inform whether to re-treat or to switch to another disease-modifying treatment.


Assuntos
Alemtuzumab/imunologia , Anticorpos Monoclonais Humanizados/imunologia , Antígeno CD52/imunologia , Esclerose Múltipla Recidivante-Remitente/imunologia , Animais , Anticorpos Monoclonais Humanizados/uso terapêutico , Anticorpos Neutralizantes/imunologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Humanos , Depleção Linfocítica/métodos , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Ratos
18.
Biotechniques ; 68(4): 185-190, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32096651

RESUMO

Aim: The humanized anti-CD52 monoclonal antibody alemtuzumab depletes lymphocytes and is currently used to treat relapsing multiple sclerosis. During treatment, anti-alemtuzumab antibodies may develop and reduce effective lymphocyte depletion in future treatment cycles. Results: Alemtuzumab-Alexa Fluor 488 conjugate binding to the CHO-CD52 cell surface was inhibited by anti-alemtuzumab antibodies. Conclusion: In this proof-of-concept study, a CHO-CD52 cell line has been developed and used to detect the presence of anti-alemtuzumab neutralizing antibodies. This platform provides the basis of an assay for routine screening of serum for neutralizing antibodies from patients treated with alemtuzumab.


Assuntos
Alemtuzumab/imunologia , Anticorpos Neutralizantes , Técnicas Citológicas/métodos , Imunoensaio/métodos , Alemtuzumab/uso terapêutico , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Ligação Competitiva/imunologia , Antígeno CD52/imunologia , Antígeno CD52/metabolismo , Células CHO/química , Células CHO/metabolismo , Cricetulus , Fluoresceínas , Humanos , Depleção Linfocítica/métodos , Esclerose Múltipla/tratamento farmacológico , Ácidos Sulfônicos
19.
Sci Rep ; 10(1): 1860, 2020 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-32024871

RESUMO

The occurrence of anti-drug antibodies following administration of therapeutic monoclonal antibody to patients is a growing problem that is attracting attention from frontline clinicians. Ideally, an initial indicative point of care test would provide guidance to seek testing approved by the regulatory authorities. Here we describe a platform for the detection of IgG anti-drug antibodies that may provide an initial screen for all therapeutic monoclonal antibodies. Synthetic genes encoding Nanoluciferase polypeptides were inserted between the variable heavy and light domain encoding region of known antibody drugs (alemtuzumab and adalimumab) to generate recombinant single chain GloBodies, which retain the drug antibody paratopes and Nanoluciferase activity. In the presence of anti-drug antibodies, the GloBody is bound by specific IgG in the sample. These complexes are captured on immobilised Protein G and the luciferase activity determined. The amount of light generated being indicative of the anti-drug IgG antibody levels in serum. It should be possible to assemble GloBody reagents for all therapeutic monoclonal antibodies and adapt the capture phase to include additional specific isotypes. The assay has the potential to be developed for use with a drop of blood allowing initial pre-screening in a point of care setting.


Assuntos
Anticorpos Monoclonais/imunologia , Biotecnologia/métodos , Imunoensaio/métodos , Preparações Farmacêuticas/metabolismo , Sítios de Ligação de Anticorpos/imunologia , Humanos , Imunoglobulina G/imunologia , Luciferases/imunologia , Sistemas Automatizados de Assistência Junto ao Leito
20.
Immunology ; 154(2): 253-260, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29247512

RESUMO

Alemtuzumab is a lymphocyte-depleting antibody and one of the most effective treatments for relapsing multiple sclerosis. However, it also causes loss of immune-tolerance leading to secondary autoimmunity and marked anti-drug antibody responses. Although these anti-drug responses have been reported to be of no significance, we hypothesized that they will affect the depleting capacity and treatment response in some individuals. This was found following analysis of the regulatory submission of the pivotal phase III trials, which was obtained from the European Medicines Agency. At the population level there was lack of influence of 'ever-positive' alemtuzumab-specific antibody responses on lymphocyte depletion, clinical efficacy and adverse effects during the 2-year trial. This was not surprising as no one before the first infusion, and only 0·6% of people before the second-infusion, had pre-infusion, neutralizing antibodies (NAbs). However, at the individual level, NAbs led to poor lymphocyte depletion. Importantly, it was evident that 31% of people had NAbs and 75% had binding antibodies at the end of treatment-cycle 2, which suggests that problems may occur in people requiring additional alemtuzumab cycles. In addition, we also identified individuals, following 'post-marketing' alemtuzumab use, whose lymphocyte level was never effectively depleted after the first infusion cycle. Hence, although alemtuzumab depletes lymphocytes in most individuals, some people fail to deplete/deplete poorly, probably due to biological-response variation and NAbs, and this may lead to treatment failure. Monitoring depletion following infusion and assessment of the neutralizing response before re-infusion may help inform the decision to retreat or switch therapy to limit treatment failure.


Assuntos
Alemtuzumab/farmacologia , Depleção Linfocítica , Esclerose Múltipla/imunologia , Alemtuzumab/uso terapêutico , Anticorpos Neutralizantes/farmacologia , Anticorpos Neutralizantes/uso terapêutico , Humanos , Depleção Linfocítica/métodos , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/metabolismo , Falha de Tratamento , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA