Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
N Engl J Med ; 384(14): 1323-1334, 2021 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-33826820

RESUMO

BACKGROUND: Cold agglutinin disease is a rare autoimmune hemolytic anemia characterized by hemolysis that is caused by activation of the classic complement pathway. Sutimlimab, a humanized monoclonal antibody, selectively targets the C1s protein, a C1 complex serine protease responsible for activating this pathway. METHODS: We conducted a 26-week multicenter, open-label, single-group study to assess the efficacy and safety of intravenous sutimlimab in patients with cold agglutinin disease and a recent history of transfusion. The composite primary end point was a normalization of the hemoglobin level to 12 g or more per deciliter or an increase in the hemoglobin level of 2 g or more per deciliter from baseline, without red-cell transfusion or medications prohibited by the protocol. RESULTS: A total of 24 patients were enrolled and received at least one dose of sutimlimab; 13 patients (54%) met the criteria for the composite primary end point. The least-squares mean increase in hemoglobin level was 2.6 g per deciliter at the time of treatment assessment (weeks 23, 25, and 26). A mean hemoglobin level of more than 11 g per deciliter was maintained in patients from week 3 through the end of the study period. The mean bilirubin levels normalized by week 3. A total of 17 patients (71%) did not receive a transfusion from week 5 through week 26. Clinically meaningful reductions in fatigue were observed by week 1 and were maintained throughout the study. Activity in the classic complement pathway was rapidly inhibited, as assessed by a functional assay. Increased hemoglobin levels, reduced bilirubin levels, and reduced fatigue coincided with inhibition of the classic complement pathway. At least one adverse event occurred during the treatment period in 22 patients (92%). Seven patients (29%) had at least one serious adverse event, none of which were determined by the investigators to be related to sutimlimab. No meningococcal infections occurred. CONCLUSIONS: In patients with cold agglutinin disease who received sutimlimab, selective upstream inhibition of activity in the classic complement pathway rapidly halted hemolysis, increased hemoglobin levels, and reduced fatigue. (Funded by Sanofi; CARDINAL ClinicalTrials.gov number, NCT03347396.).


Assuntos
Anemia Hemolítica Autoimune/tratamento farmacológico , Anticorpos Monoclonais Humanizados/uso terapêutico , Complemento C1s/antagonistas & inibidores , Idoso , Idoso de 80 Anos ou mais , Anemia Hemolítica Autoimune/sangue , Anemia Hemolítica Autoimune/complicações , Anemia Hemolítica Autoimune/terapia , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/farmacologia , Transfusão de Sangue , Fadiga/tratamento farmacológico , Fadiga/etiologia , Feminino , Hemoglobinas/análise , Hemólise/efeitos dos fármacos , Humanos , Masculino , Pessoa de Meia-Idade , Qualidade de Vida
3.
Mol Immunol ; 126: 8-13, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32717572

RESUMO

The serpin, C1-inhibitor (also known as SERPING1), plays a vital anti-inflammatory role in the body by controlling pro-inflammatory pathways such as complement and coagulation. The inhibitor's action is enhanced in the presence of polyanionic cofactors, such as heparin and polyphosphate, by increasing the rate of association with key enzymes such as C1s of the classical pathway of complement. The cofactor binding site of the serpin has never been mapped. Here we show that residues Lys284, Lys285 and Arg287 of C1-inhibitor play key roles in binding heparin and delivering the rate enhancement seen in the presence of polyanions and thus most likely represent the key cofactor binding residues for the serpin. We also show that simultaneous binding of the anion binding site of C1s by the polyanion is required to deliver the rate enhancement. Finally, we have shown that it is unlikely that the two positively charged zones of C1-inhibitor and C1s interact in the encounter complex between molecules as ablation of the charged zones did not in itself deliver a rate enhancement as might have been expected if the zones interacted. These insights provide crucial information as to the mechanism of action of this key serpin in the presence and absence of cofactor molecules.


Assuntos
Proteína Inibidora do Complemento C1/metabolismo , Complemento C1s/antagonistas & inibidores , Polímeros/metabolismo , Sítios de Ligação/genética , Proteína Inibidora do Complemento C1/genética , Proteína Inibidora do Complemento C1/isolamento & purificação , Complemento C1s/metabolismo , Heparina/metabolismo , Mutagênese Sítio-Dirigida , Mutação , Polieletrólitos , Polifosfatos/metabolismo , Ligação Proteica/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo
4.
Molecules ; 24(20)2019 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-31600984

RESUMO

The complement system is associated with various diseases such as inflammation or auto-immune diseases. Complement-targeted drugs could provide novel therapeutic intervention against the above diseases. C1s, a serine protease, plays an important role in the CS and could be an attractive target since it blocks the system at an early stage of the complement cascade. Designing C1 inhibitors is particularly challenging since known inhibitors are restricted to a narrow bioactive chemical space in addition selectivity over other serine proteases is an important requirement. The typical architecture of a small molecule inhibitor of C1s contains an amidine (or guanidine) residue, however, the discovery of non-amidine inhibitors might have high value, particularly if novel chemotypes and/or compounds displaying improved selectivity are identified. We applied various virtual screening approaches to identify C1s focused libraries that lack the amidine/guanidine functionalities, then the in silico generated libraries were evaluated by in vitro biological assays. While 3D structure-based methods were not suitable for virtual screening of C1s inhibitors, and a 2D similarity search did not lead to novel chemotypes, pharmacophore model generation allowed us to identify two novel chemotypes with submicromolar activities. In three screening rounds we tested altogether 89 compounds and identified 20 hit compounds (<10 µM activities; overall hit rate: 22.5%). The highest activity determined was 12 nM (1,2,4-triazole), while for the newly identified chemotypes (1,3-benzoxazin-4-one and thieno[2,3-d][1,3]oxazin-4-one) it was 241 nM and 549 nM, respectively.


Assuntos
Complemento C1s/antagonistas & inibidores , Complemento C1s/química , Desenho de Fármacos , Descoberta de Drogas , Modelos Moleculares , Desenvolvimento de Medicamentos , Descoberta de Drogas/métodos , Estrutura Molecular , Relação Quantitativa Estrutura-Atividade , Bibliotecas de Moléculas Pequenas
5.
Transplantation ; 103(9): 1763-1775, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30946220

RESUMO

Complement plays important roles in both ischemia-reperfusion injury (IRI) and antibody-mediated rejection (AMR) of solid organ allografts. One approach to possibly improve outcomes after transplantation is the use of C1 inhibitor (C1-INH), which blocks the first step in both the classical and lectin pathways of complement activation and also inhibits the contact, coagulation, and kinin systems. C1-INH can also directly block leukocyte-endothelial cell adhesion. C1-INH contrasts with eculizumab and other distal inhibitors, which do not affect C4b or C3b deposition or noncomplement pathways. Authors of reports on trials in kidney transplant recipients have suggested that C1-INH treatment may reduce IRI and delayed graft function, based on decreased requirements for dialysis in the first month after transplantation. This effect was particularly marked with grafts with Kidney Disease Profile Index ≥ 85. Other clinical studies and models suggest that C1-INH may decrease sensitization and donor-specific antibody production and might improve outcomes in AMR, including in patients who are refractory to other modalities. However, the studies have been small and often only single-center. This article reviews clinical data and ongoing trials with C1-INH in transplant recipients, compares the results with those of other complement inhibitors, and summarizes potentially productive directions for future research.


Assuntos
Ativação do Complemento/efeitos dos fármacos , Proteína Inibidora do Complemento C1/uso terapêutico , Complemento C1s/antagonistas & inibidores , Inativadores do Complemento/uso terapêutico , Função Retardada do Enxerto/prevenção & controle , Rejeição de Enxerto/prevenção & controle , Transplante de Órgãos , Traumatismo por Reperfusão/prevenção & controle , Aloenxertos , Animais , Proteína Inibidora do Complemento C1/efeitos adversos , Complemento C1s/imunologia , Inativadores do Complemento/efeitos adversos , Função Retardada do Enxerto/imunologia , Rejeição de Enxerto/imunologia , Sobrevivência de Enxerto/efeitos dos fármacos , Humanos , Transplante de Órgãos/efeitos adversos , Traumatismo por Reperfusão/imunologia , Fatores de Risco , Resultado do Tratamento
6.
Nucleic Acid Ther ; 29(2): 82-91, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30817230

RESUMO

Kallikrein is the key contact system mediator responsible for the conversion of high-molecular-weight kininogen into the inflammatory vasodilator peptide bradykinin, a process regulated by C1-esterase inhibitor (C1-INH). In hereditary angioedema (HAE), genetic mutations result in deficient or dysfunctional C1-INH and dysregulation of the contact system leading to recurrent, sometimes fatal, angioedema attacks. IONIS-PKKRx is a second-generation 2'-O-(2-methoxyethyl)-modified chimeric antisense oligonucleotide, designed to bind and selectively reduce prekallikrein (PKK) mRNA in the liver. IONIS-PKKRx demonstrated dose-dependent reduction of human prekallikrein hepatic mRNA and plasma protein in transgenic mice and dose- and time-dependent reductions of plasma PKK in Cynomolgus monkeys. Similar dose-dependent reductions of plasma PKK levels were observed in healthy human volunteers accompanied by decreases in bradykinin generation capacity with an acceptable safety and tolerability profile. These results highlight a novel and specific approach to target PKK for the treatment of HAE and other diseases involving contact system activation and overproduction of bradykinin.


Assuntos
Angioedemas Hereditários/terapia , Bradicinina/genética , Complemento C1s/genética , Pré-Calicreína/genética , Angioedemas Hereditários/sangue , Angioedemas Hereditários/genética , Animais , Animais Geneticamente Modificados/sangue , Bradicinina/sangue , Proteína Inibidora do Complemento C1/farmacologia , Complemento C1s/antagonistas & inibidores , Relação Dose-Resposta a Droga , Voluntários Saudáveis , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Macaca fascicularis/sangue , Camundongos , Oligodesoxirribonucleotídeos Antissenso/genética , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Pré-Calicreína/antagonistas & inibidores
7.
J Immunol ; 202(4): 1200-1209, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30635392

RESUMO

The classical pathway of complement (CP) can mediate C3 opsonization of Ags responsible for the costimulation and activation of cognate B lymphocytes. In this manner, the complement system acts as a bridge between the innate and adaptive immune systems critical for establishing a humoral response. However, aberrant complement activation is often observed in autoimmune diseases in which C3 deposition on self-antigens may serve to activate self-reactive B cell clones. In this study, we use BIVV009 (Sutimlimab), a clinical stage, humanized mAb that specifically inhibits the CP-specific serine protease C1s to evaluate the impact of upstream CP antagonism on activation and proliferation of normal and autoimmune human B cells. We report that BIVV009 significantly inhibited complement-mediated activation and proliferation of primary human B cells. Strikingly, CP antagonism suppressed human Ig-induced activation of B cells derived from patients with rheumatoid arthritis. These results suggest that clinical use of CP inhibitors in autoimmune patients may not only block complement-mediated tissue damage, but may also prevent the long-term activation of autoimmune B cells and the production of autoantibodies that contribute to the underlying pathologic condition of these diseases.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Artrite Reumatoide/tratamento farmacológico , Linfócitos B/efeitos dos fármacos , Complemento C1s/antagonistas & inibidores , Artrite Reumatoide/imunologia , Artrite Reumatoide/patologia , Linfócitos B/imunologia , Linfócitos B/patologia , Proliferação de Células/efeitos dos fármacos , Ativação do Complemento/efeitos dos fármacos , Complemento C1s/imunologia , Humanos
8.
Blood ; 133(9): 893-901, 2019 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-30559259

RESUMO

Cold agglutinin disease is a difficult-to-treat autoimmune hemolytic anemia in which immunoglobulin M antibodies bind to erythrocytes and fix complement, resulting in predominantly extravascular hemolysis. This trial tested the hypothesis that the anti-C1s antibody sutimlimab would ameliorate hemolytic anemia. Ten patients with cold agglutinin disease participated in the phase 1b component of a first-in-human trial. Patients received a test dose of 10-mg/kg sutimlimab followed by a full dose of 60 mg/kg 1 to 4 days later and 3 additional weekly doses of 60 mg/kg. All infusions were well tolerated without premedication. No drug-related serious adverse events were observed. Seven of 10 patients with cold agglutinin disease responded with a hemoglobin increase >2 g/dL. Sutimlimab rapidly increased hemoglobin levels by a median of 1.6 g/dL within the first week, and by a median of 3.9 g/dL (interquartile range, 1.3-4.5 g/dL; 95% confidence interval, 2.1-4.5) within 6 weeks (P = .005). Sutimlimab rapidly abrogated extravascular hemolysis, normalizing bilirubin levels within 24 hours in most patients and normalizing haptoglobin levels in 4 patients within 1 week. Hemolytic anemia recurred when drug levels were cleared from the circulation 3 to 4 weeks after the last dose of sutimlimab. Reexposure to sutimlimab in a named patient program recapitulated the control of hemolytic anemia. All 6 previously transfused patients became transfusion-free during treatment. Sutimlimab was safe, well tolerated, and rapidly stopped C1s complement-mediated hemolysis in patients with cold agglutinin disease, significantly increasing hemoglobin levels and precluding the need for transfusions. This trial was registered at www.clinicaltrials.gov as #NCT02502903.


Assuntos
Anemia Hemolítica Autoimune/tratamento farmacológico , Anemia Hemolítica/prevenção & controle , Anticorpos Monoclonais Humanizados/uso terapêutico , Complemento C1s/antagonistas & inibidores , Hemólise/efeitos dos fármacos , Índice de Gravidade de Doença , Idoso , Anemia Hemolítica/etiologia , Anemia Hemolítica Autoimune/complicações , Complemento C1s/imunologia , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Estudos Prospectivos
9.
Clin Pharmacol Ther ; 104(4): 655-663, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29737533

RESUMO

Aberrant activation of the classical complement pathway is the common underlying pathophysiology of orphan diseases such as bullous pemphigoid, antibody-mediated rejection of organ transplants, cold agglutinin disease, and warm autoimmune hemolytic anemia. Therapeutic options for these complement-mediated disorders are limited and sutimlimab, a humanized monoclonal antibody directed against complement factor C1s, may be potentially useful for inhibition of the classical complement pathway. A phase I, first-in-human, double-blind, randomized, placebo-controlled, dose-escalation trial of single and multiple doses of sutimlimab or placebo was conducted in 64 volunteers to evaluate safety, tolerability, pharmacokinetic, and pharmacodynamic profiles. Single and multiple infusions of sutimlimab were well tolerated without any safety concerns. sutimlimab exhibited a steep concentration-effect relationship with a Hill coefficient of 2.4, and an IC90 of 15.5 µg/mL. This study establishes the foundation for using sutimlimab as a highly selective inhibitor of the classical complement pathway in different diseases.


Assuntos
Anticorpos Monoclonais Humanizados/administração & dosagem , Complemento C1s/antagonistas & inibidores , Inativadores do Complemento/administração & dosagem , Via Clássica do Complemento/efeitos dos fármacos , Adulto , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/farmacocinética , Áustria , Complemento C1s/imunologia , Inativadores do Complemento/efeitos adversos , Inativadores do Complemento/farmacocinética , Relação Dose-Resposta a Droga , Método Duplo-Cego , Esquema de Medicação , Feminino , Voluntários Saudáveis , Humanos , Infusões Intravenosas , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento , Adulto Jovem
11.
Transplantation ; 101(10): 2410-2418, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28926521

RESUMO

BACKGROUND: Complement may play a key role in antibody-mediated rejection. A promising therapeutic approach may be classical pathway (CP) inhibition at the level of early component C1. METHODS: In this first-in-human, double-blind, randomized placebo-controlled phase 1 trial, we evaluated the safety and complement inhibitory effect of TNT009, a humanized monoclonal anti-C1s antibody. Sixty-four adult healthy volunteers received either single (n = 48; 7 consecutive cohorts, 0.3-100 mg/kg) or 4 weekly infusions (n = 16; 2 consecutive cohorts, 30 and 60 mg/kg per infusion) of TNT009 or placebo. To assess the effect of treatment on complement activity, sera from dosed subjects were analyzed in a CP activation assay evaluating C3d deposition on HLA-coated microbeads spiked with alloantibodies. RESULTS: Single doses of TNT009 at 3 to 100 mg/kg uniformly and profoundly inhibited HLA antibody-mediated C3d deposition (≥86% after 60 minutes), whereby the duration of CP inhibition (2-14 days) was dose-dependent. Four weekly doses persistently blocked complement for 5 to 6 weeks. Ex vivo serum CP activity was profoundly inhibited when TNT009 concentrations exceeded 20 µg/mL. Infusions were well tolerated without serious or severe adverse events. CONCLUSIONS: Treatment with TNT009 was safe and potently inhibited CP activity. Future studies in patients are required to assess the potential of TNT009 for preventing or treating antibody-mediated rejection.


Assuntos
Anticorpos Monoclonais Humanizados/administração & dosagem , Ativação do Complemento/efeitos dos fármacos , Complemento C1s/antagonistas & inibidores , Inativadores do Complemento/administração & dosagem , Antígenos HLA/imunologia , Adulto , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/sangue , Anticorpos Monoclonais Humanizados/uso terapêutico , Complemento C1s/imunologia , Inativadores do Complemento/efeitos adversos , Inativadores do Complemento/sangue , Método Duplo-Cego , Esquema de Medicação , Feminino , Voluntários Saudáveis , Humanos , Infusões Intravenosas , Masculino , Fatores de Tempo , Resultado do Tratamento
12.
J Immunol ; 198(12): 4728-4737, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28484054

RESUMO

The complement system is a front-line defense system that opsonizes and lyses invading pathogens. To survive, microbes exposed to serum must evade the complement response. To achieve this, many pathogens recruit soluble human complement regulators to their surfaces and hijack their regulatory function for protection from complement activation. C1 esterase inhibitor (C1-INH) is a soluble regulator of complement activation that negatively regulates the classical and lectin pathways of complement to protect human tissue from aberrant activation. In this article, we show that Plasmodium falciparum merozoites, the invasive form of blood stage malaria parasites, actively recruit C1-INH to their surfaces when exposed to human serum. We identified PfMSP3.1, a member of the merozoite surface protein 3 family of merozoite surface proteins, as the direct interaction partner. When bound to the merozoite surface, C1-INH retains its ability to complex with and inhibit C1s, MASP1, and MASP2, the activating proteases of the complement cascade. P. falciparum merozoites that lack PfMSP3.1 showed a marked reduction in C1-INH recruitment and increased C3b deposition on their surfaces. However, these ΔPfMSP3.1 merozoites exhibit enhanced invasion of RBCs in the presence of active complement. This study characterizes an immune-evasion strategy used by malaria parasites and highlights the complex relationship between merozoites and the complement system.


Assuntos
Antígenos de Protozoários/metabolismo , Ativação do Complemento , Proteína Inibidora do Complemento C1/metabolismo , Evasão da Resposta Imune , Proteínas de Membrana/metabolismo , Merozoítos/imunologia , Plasmodium falciparum/imunologia , Antígenos de Protozoários/imunologia , Proteína Inibidora do Complemento C1/genética , Complemento C1s/antagonistas & inibidores , Complemento C1s/imunologia , Complemento C1s/metabolismo , Eritrócitos/parasitologia , Humanos , Serina Proteases Associadas a Proteína de Ligação a Manose/antagonistas & inibidores , Serina Proteases Associadas a Proteína de Ligação a Manose/imunologia , Serina Proteases Associadas a Proteína de Ligação a Manose/metabolismo , Proteínas de Membrana/imunologia , Merozoítos/química , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium falciparum/metabolismo
13.
Clin Immunol ; 163: 84-90, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26769276

RESUMO

Complement activation contributes to inflammation in many diseases, yet it also supports physiologic apoptotic cells (AC) clearance and its downstream immunosuppressive effects. The roles of individual complement components in AC phagocytosis have been difficult to dissect with artificially depleted sera. Using human in vitro systems and the novel antibody complement C1s inhibitor TNT003, we uncoupled the role of the enzymatic activation of the classical pathway from the opsonizing role of C1q in mediating a) the phagocytosis of early and late AC, and b) the immunosuppressive capacity of early AC. We found that C1s inhibition had a small impact on the physiologic clearance of early AC, leaving their immunosuppressive properties entirely unaffected, while mainly inhibiting the phagocytosis of late apoptotic/secondary necrotic cells. Our data suggest that C1s inhibition may represent a valuable therapeutic strategy to control classical pathway activation without causing significant AC accumulation in diseases without defects in AC phagocytosis.


Assuntos
Anticorpos Monoclonais/farmacologia , Apoptose/efeitos dos fármacos , Ativação do Complemento/efeitos dos fármacos , Complemento C1q/efeitos dos fármacos , Complemento C1s/antagonistas & inibidores , Citocinas/efeitos dos fármacos , Citofagocitose/efeitos dos fármacos , Tolerância Imunológica/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Apoptose/imunologia , Ativação do Complemento/imunologia , Complemento C1q/imunologia , Complemento C1q/metabolismo , Complemento C1s/metabolismo , Complemento C3b/efeitos dos fármacos , Complemento C3b/imunologia , Complemento C3b/metabolismo , Citocinas/imunologia , Citofagocitose/imunologia , Humanos , Técnicas In Vitro , Interleucina-1beta/efeitos dos fármacos , Interleucina-1beta/imunologia , Interleucina-6/imunologia , Células Jurkat , Macrófagos/imunologia , Fagocitose/efeitos dos fármacos , Fagocitose/imunologia , Fator de Necrose Tumoral alfa/efeitos dos fármacos , Fator de Necrose Tumoral alfa/imunologia
15.
Blood ; 123(26): 4010-2, 2014 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-24970929

RESUMO

In this issue of Blood, Shi and coworkers show that TNT003, a mouse monoclonal antibody targeting complement protein C1s, prevents induction of in vitro hemolysis by cold agglutinins (CA). If successfully transferred into the clinical setting by further studies, these findings may result in a novel therapeutic principle for a frequently difficult problem.


Assuntos
Anemia Hemolítica Autoimune/tratamento farmacológico , Anticorpos Monoclonais Murinos/farmacologia , Ativação do Complemento/efeitos dos fármacos , Complemento C1s/antagonistas & inibidores , Serina Proteases/sangue , Inibidores de Serina Proteinase/farmacologia , Animais , Feminino , Humanos , Masculino
16.
Blood ; 123(26): 4015-22, 2014 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-24695853

RESUMO

Activation of the classical pathway (CP) of complement is often associated with autoimmune disorders in which disease pathology is linked to the presence of an autoantibody. One such disorder is cold agglutinin disease (CAD), an autoimmune hemolytic anemia in which autoantibodies (cold agglutinins) bind to red blood cells (RBCs) at low temperatures. Anemia occurs as a result of autoantibody-mediated CP activation on the surface of the erythrocyte, leading to the deposition of complement opsonins that drive extravascular hemolysis in the liver. Here we test the effects of TNT003, a mouse monoclonal antibody targeting the CP-specific serine protease C1s, on CP activity induced by cold agglutinins on human RBCs. We collected 40 individual CAD patient samples and showed that TNT003 prevented cold agglutinin-mediated deposition of complement opsonins that promote phagocytosis of RBCs. Furthermore, we show that by preventing CP activation, TNT003 also prevents cold agglutinin-driven generation of anaphylatoxins. Finally, we provide evidence that CP activity in CAD patients terminates prior to activation of the terminal cascade, supporting the hypothesis that the primary route of RBC destruction in these patients occurs via extravascular hemolysis. Our results support the development of a CP inhibitor for the treatment of CAD.


Assuntos
Anemia Hemolítica Autoimune/tratamento farmacológico , Anticorpos Monoclonais Murinos/farmacologia , Ativação do Complemento/efeitos dos fármacos , Complemento C1s/antagonistas & inibidores , Serina Proteases/sangue , Inibidores de Serina Proteinase/farmacologia , Idoso , Anafilatoxinas/metabolismo , Anemia Hemolítica Autoimune/enzimologia , Anemia Hemolítica Autoimune/patologia , Animais , Linhagem Celular Tumoral , Complemento C1s/metabolismo , Feminino , Hemólise/efeitos dos fármacos , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade
17.
J Allergy Clin Immunol Pract ; 2(1): 77-84, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24565773

RESUMO

BACKGROUND: Nanofiltered C1 inhibitor (human) is approved in the United States for routine prophylaxis of angioedema attacks in patients with hereditary angioedema, a rare disease caused by a deficiency of functional C1 inhibitor. OBJECTIVE: To assess the safety of escalating doses of nanofiltered C1 inhibitor (human) in patients who were not adequately controlled on the indicated dose (1000 U every 3 or 4 days). METHODS: Eligible patients had >1 attack/month over the 3 months before the trial. Doses were escalated to 1500 U every 3 or 4 days for 12 weeks, at which point, the patients were evaluated. If treatment was successful (≤1 attack/mo) or at the investigator's discretion, the patients entered a 3-month follow-up period. The patients with an average of >1 attack/month were eligible for further escalation to 2000 U and then 2500 U. RESULTS: Twenty patients started at 1500 U; 13 were escalated to 2000 U, and 12 were escalated to 2500 U. Eighteen patients reported adverse events. Two patients reported 4 serious adverse events (cerebral cystic hygroma, laryngeal angioedema attack, anemia, and bile duct stone) that were considered by investigators to be unrelated to treatment. Notably, there were no systemic thrombotic events or discontinuations due to adverse events. Fourteen patients were treated successfully (70%), continued to the follow-up period at the investigator's discretion, or experienced a reduction in attacks of >1.0/month. CONCLUSIONS: Dose escalation of nanofiltered C1 inhibitor (human) up to 2500 U was well tolerated and reduced attack frequency in the majority of patients.


Assuntos
Angioedemas Hereditários/prevenção & controle , Proteínas Inativadoras do Complemento 1/administração & dosagem , Complemento C1s/antagonistas & inibidores , Adolescente , Adulto , Idoso , Algoritmos , Angioedemas Hereditários/diagnóstico , Angioedemas Hereditários/enzimologia , Angioedemas Hereditários/imunologia , Proteínas Inativadoras do Complemento 1/efeitos adversos , Proteína Inibidora do Complemento C1 , Complemento C1s/metabolismo , Esquema de Medicação , Cálculos da Dosagem de Medicamento , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fatores de Tempo , Resultado do Tratamento , Estados Unidos , Adulto Jovem
18.
Dev Comp Immunol ; 42(2): 197-210, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24056051

RESUMO

C1 inhibitor (C1Inh), a member of serpin superfamily, is a crucial regulator of the activation of various plasmatic cascades associated with immunity and inflammation. This study describes the identification and characterization of a C1Inh gene from rock bream Oplegnathus fasciatus (OfC1Inh) at structural, expressional and functional levels. The cDNA-(2245bp) and corresponding gDNA-sequences (5.2kbp) of OfC1Inh were isolated from rock bream transcriptome- and BAC-libraries, respectively. Predicted amino acid sequence of OfC1Inh revealed a two-domain architecture composed of an N-terminal region with two Ig-like domains and a C-terminal region with a serpin domain. Tertiary model of OfC1Inh disclosed its active site topology. In the multi-exonic genomic arrangement of OfC1Inh, it consisted of eleven exons disjoined by ten introns as observed in few other fish homologs. Our comparative analysis indicated that the teleostean C1Inhs were distinct from their non-teleostean vertebrate counterparts in terms of their (1) extended N-terminal domains, (2) evolutionary divergence and (3) exon-intron distribution. The OfC1Inh had a TATA-deficient promoter with a putative initiator element, and two tandemly arranged downstream promoter elements. Several components associated with the immune and inflammatory transcriptional activation were also predicted to exist in 5' flanking region of OfC1Inh. The exclusive mRNA levels in liver and moderate levels in extra-hepatic tissues intimated the diversified importance of OfC1Inh in rock bream physiology. We also provide an evidence for the involvement of OfC1Inh in immune balance, based on its modulated transcription upon different PAMP (lipopolysaccharide and poly I:C)- or pathogen (Streptococcus iniae and rock bream irido virus)-challenges. A recombinantly expressed fusion protein [(r)OfC1Inh] was employed in demonstrating the anti-protease function of OfC1Inh. The (r)OfC1Inh exhibited detectable inhibitory activity against C1 esterase and thrombin, where the anti-C1 esterase role was shown to be potentiated by heparin. Taken together, the results of this study provide the first line of evidence for the possible involvement of a teleostean C1Inh in fish immunity, based on its expressional response(s) and inhibitory properties against two enzymes involved in biological cascades.


Assuntos
Proteína Inibidora do Complemento C1/genética , Proteína Inibidora do Complemento C1/imunologia , Complemento C1s/antagonistas & inibidores , Peixes/imunologia , Trombina/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Sequência de Bases , Proteínas de Peixes/genética , Proteínas de Peixes/imunologia , Peixes/genética , Perfilação da Expressão Gênica , Heparina/farmacologia , Inflamação/imunologia , Interferon Tipo I/imunologia , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Estrutura Terciária de Proteína , RNA Mensageiro/biossíntese , Proteínas Recombinantes/imunologia , Alinhamento de Sequência , Análise de Sequência de DNA
19.
Immunobiology ; 218(8): 1041-8, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23434433

RESUMO

Disregulation of complement activation plays a critical role in numerous inflammatory diseases and therefore, inhibition of the complement pathway is of great therapeutic interest. In the classical complement pathway, immune complexes formed by IgM, IgG1, IgG2 and IgG3 antibodies result in the activation of the C1s protease that in turn cleaves C4 and then C4-bound-C2 yielding the proteolytic fragments C4b and C2a which associate to form a C3 convertase enzyme. We report here the engineering of a potent human antibody inhibitor of C1s protease activity. Phage panning of a very large synthetic (F(AB)) antibody fragment library using a truncated version of C1s, comprising the second CCP domain and serine protease domain (CCP2-SP) and expressed in insect cells, resulted in the isolation of a F(AB) that inhibited the catalytic activity of C1s. An affinity matured variant of the F(AB) format antibody displaying subnanomolar K(D) for C1s was shown to exhibit >80% inhibition of C2 processing at a 5:1 antibody:C1s molar ratio. We show that this engineered antibody, D.35, displays potent inhibition of complement deposition and lysis of Ramos cells by the anti-CD20 therapeutic antibody rituximab relative to the approved, but less-specific, human plasma-derived C1-inhibitor (CINRYZE). C1s inhibitory antibodies should be useful for delineating the role of the classical pathway in disease models and may hold promise as therapeutic agents.


Assuntos
Anticorpos/imunologia , Ativação do Complemento/imunologia , Complemento C1s/antagonistas & inibidores , Fragmentos Fab das Imunoglobulinas/imunologia , Complemento C1s/imunologia , Complemento C1s/metabolismo , Complemento C4/imunologia , Humanos
20.
Pharmacotherapy ; 32(10): 902-9, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23033229

RESUMO

STUDY OBJECTIVE: To investigate reports of thrombotic events associated with the use of C1 esterase inhibitor products in patients with hereditary angioedema in the United States. DESIGN: Retrospective data mining analysis. SOURCE: The United States Food and Drug Administration (FDA) adverse event reporting system (AERS) database. MEASUREMENTS AND MAIN RESULTS: Case reports of C1 esterase inhibitor products, thrombotic events, and C1 esterase inhibitor product-associated thrombotic events (i.e., combination cases) were extracted from the AERS database, using the time frames of each respective product's FDA approval date through the second quarter of 2011. Bayesian statistical methodology within the neural network architecture was implemented to identify potential signals of a drug-associated adverse event. A potential signal is generated when the lower limit of the 95% 2-sided confidence interval of the information component, denoted by IC025 , is greater than zero. This suggests that the particular drug-associated adverse event was reported to the database more often than statistically expected from reports available in the database. Ten combination cases of thrombotic events associated with the use of one C1 esterase inhibitor product (Cinryze) were identified in patients with hereditary angioedema. A potential signal demonstrated by an IC025 value greater than zero (IC025 = 2.91) was generated for these combination cases. CONCLUSION: The extracted cases from the AERS indicate continuing reports of thrombotic events associated with the use of one C1 esterase inhibitor product among patients with hereditary angioedema. The AERS is incapable of establishing a causal link and detecting the true frequency of an adverse event associated with a drug; however, potential signals of C1 esterase inhibitor product-associated thrombotic events among patients with hereditary angioedema were identified in the extracted combination cases.


Assuntos
Angioedemas Hereditários/tratamento farmacológico , Complemento C1s/antagonistas & inibidores , Inativadores do Complemento/efeitos adversos , Trombose/induzido quimicamente , Adolescente , Adulto , Sistemas de Notificação de Reações Adversas a Medicamentos , Idoso , Teorema de Bayes , Criança , Proteínas Inativadoras do Complemento 1/efeitos adversos , Proteínas Inativadoras do Complemento 1/uso terapêutico , Proteína Inibidora do Complemento C1 , Inativadores do Complemento/uso terapêutico , Mineração de Dados , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Redes Neurais de Computação , Peptídeos/efeitos adversos , Peptídeos/uso terapêutico , Estudos Retrospectivos , Estados Unidos , United States Food and Drug Administration , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA