Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
J Cell Mol Med ; 28(13): e18470, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38963257

RESUMO

Recombinant antibodies (Abs) are an integral modality for the treatment of multiple tumour malignancies. Since the Food and Drug Administration (FDA) approval of rituximab as the first monoclonal antibody (mAb) for cancer treatment, several mAbs and antibody (Ab)-based therapies have been approved for the treatment of solid tumour malignancies and other cancers. These Abs function by either blocking oncogenic pathways or angiogenesis, modulating immune response, or by delivering a conjugated drug. The use of Ab-based therapy in cancer patients who could benefit from the treatment, however, is still limited by associated toxicity profiles which may stem from biological features and processes related to target binding, alongside biochemical and/or biophysical characteristics of the therapeutic Ab. A significant immune-related adverse event (irAE) associated with Ab-based therapies is cytokine release syndrome (CRS), characterized by the development of fever, rash and even marked, life-threatening hypotension, and acute inflammation with secondary to systemic uncontrolled increase in a range of pro-inflammatory cytokines. Here, we review irAEs associated with specific classes of approved, Ab-based novel cancer immunotherapeutics, namely immune checkpoint (IC)-targeting Abs, bispecific Abs (BsAbs) and Ab-drug-conjugates (ADCs), highlighting the significance of harmonization in preclinical assay development for safety assessment of Ab-based biotherapeutics as an approach to support and refine clinical translation.


Assuntos
Neoplasias , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Anticorpos Biespecíficos/efeitos adversos , Anticorpos Biespecíficos/uso terapêutico , Imunoterapia/efeitos adversos , Imunoterapia/métodos , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais/efeitos adversos , Antineoplásicos Imunológicos/efeitos adversos , Antineoplásicos Imunológicos/uso terapêutico
2.
J Transl Med ; 22(1): 416, 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38698408

RESUMO

One of the most challenging aspects of developing advanced cell therapy products (CTPs) is defining the mechanism of action (MOA), potency and efficacy of the product. This perspective examines these concepts and presents helpful ways to think about them through the lens of metrology. A logical framework for thinking about MOA, potency and efficacy is presented that is consistent with the existing regulatory guidelines, but also accommodates what has been learned from the 27 US FDA-approved CTPs. Available information regarding MOA, potency and efficacy for the 27 FDA-approved CTPs is reviewed to provide background and perspective. Potency process and efficacy process charts are introduced to clarify and illustrate the relationships between six key concepts: MOA, potency, potency test, efficacy, efficacy endpoint and efficacy endpoint test. Careful consideration of the meaning of these terms makes it easier to discuss the challenges of correlating potency test results with clinical outcomes and to understand how the relationships between the concepts can be misunderstood during development and clinical trials. Examples of how a product can be "potent but not efficacious" or "not potent but efficacious" are presented. Two example applications of the framework compare how MOA is assessed in cell cultures, animal models and human clinical trials and reveals the challenge of establishing MOA in humans. Lastly, important considerations for the development of potency tests for a CTP are discussed. These perspectives can help product developers set appropriate expectations for understanding a product's MOA and potency, avoid unrealistic assumptions and improve communication among team members during the development of CTPs.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Humanos , Terapia Baseada em Transplante de Células e Tecidos/métodos , Animais , Resultado do Tratamento , United States Food and Drug Administration , Estados Unidos , Ensaios Clínicos como Assunto
3.
FASEB J ; 37(6): e22995, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37219526

RESUMO

Immuno-oncology (IO)-based therapies such as checkpoint inhibitors, bi-specific antibodies, and CAR-T-cell therapies have shown significant success in the treatment of several cancer indications. However, these therapies can result in the development of severe adverse events, including cytokine release syndrome (CRS). Currently, there is a paucity of in vivo models that can evaluate dose-response relationships for both tumor control and CRS-related safety issues. We tested an in vivo PBMC humanized mouse model to assess both treatment efficacy against specific tumors and the concurrent cytokine release profiles for individual human donors after treatment with a CD19xCD3 bispecific T-cell engager (BiTE). Using this model, we evaluated tumor burden, T-cell activation, and cytokine release in response to bispecific T-cell-engaging antibody in humanized mice generated with different PBMC donors. The results show that PBMC engrafted NOD-scid Il2rgnull mice lacking expression of mouse MHC class I and II (NSG-MHC-DKO mice) and implanted with a tumor xenograft predict both efficacy for tumor control by CD19xCD3 BiTE and stimulated cytokine release. Moreover, our findings indicate that this PBMC-engrafted model captures variability among donors for tumor control and cytokine release following treatment. Tumor control and cytokine release were reproducible for the same PBMC donor in separate experiments. The PBMC humanized mouse model described here is a sensitive and reproducible platform that identifies specific patient/cancer/therapy combinations for treatment efficacy and development of complications.


Assuntos
Leucócitos Mononucleares , Linfócitos T , Humanos , Animais , Camundongos , Camundongos Endogâmicos NOD , Resultado do Tratamento , Síndrome da Liberação de Citocina , Citocinas , Modelos Animais de Doenças , Camundongos Knockout , Camundongos SCID
4.
J Immunol ; 198(10): 3989-3998, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28320831

RESUMO

Staphylococcal enterotoxin B (SEB) is a bacterial superantigen that binds the receptors in the APC/T cell synapse and causes increased proliferation of T cells and a cytokine storm syndrome in vivo. Exposure to the toxin can be lethal and cause significant pathology in humans. The lack of effective therapies for SEB exposure remains an area of concern, particularly in scenarios of acute mass casualties. We hypothesized that blockade of the T cell costimulatory signal by the CTLA4-Ig synthetic protein (abatacept) could prevent SEB-dependent pathology. In this article, we demonstrate mice treated with a single dose of abatacept 8 h post SEB exposure had reduced pathology compared with control SEB-exposed mice. SEB-exposed mice showed significant reductions in body weight between days 4 and 9, whereas mice exposed to SEB and also treated with abatacept showed no weight loss for the duration of the study, suggesting therapeutic mitigation of SEB-induced morbidity. Histopathology and magnetic resonance imaging demonstrated that SEB mediated lung damage and edema, which were absent after treatment with abatacept. Analysis of plasma and lung tissues from SEB-exposed mice treated with abatacept demonstrated significantly lower levels of IL-6 and IFN-γ (p < 0.0001), which is likely to have resulted in less pathology. In addition, exposure of human and mouse PBMCs to SEB in vitro showed a significant reduction in levels of IL-2 (p < 0.0001) after treatment with abatacept, indicating that T cell proliferation is the main target for intervention. Our findings demonstrate that abatacept is a robust and potentially credible drug to prevent toxic effects from SEB exposure.


Assuntos
Abatacepte/uso terapêutico , Células Apresentadoras de Antígenos/imunologia , Enterotoxinas/toxicidade , Infecções Estafilocócicas/tratamento farmacológico , Linfócitos T/imunologia , Abatacepte/administração & dosagem , Abatacepte/farmacologia , Animais , Peso Corporal , Citocinas/imunologia , Enterotoxinas/farmacologia , Humanos , Interferon gama/sangue , Interleucina-2/biossíntese , Interleucina-2/imunologia , Interleucina-6/sangue , Pulmão/patologia , Camundongos , Monócitos/imunologia , Infecções Estafilocócicas/imunologia
5.
FASEB J ; 29(6): 2595-602, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25746794

RESUMO

There is an urgent unmet need for human tissue bioassays to predict cytokine storm responses to biologics. Current bioassays that detect cytokine storm responses in vitro rely on endothelial cells, usually from umbilical veins or cell lines, cocultured with freshly isolated peripheral blood mononuclear cells (PBMCs) from healthy adult volunteers. These assays therefore comprise cells from 2 separate donors and carry the disadvantage of mismatched tissues and lack the advantage of personalized medicine. Current assays also do not fully delineate mild (such as Campath) and severe (such as TGN1412) cytokine storm-inducing drugs. Here, we report a novel bioassay where endothelial cells grown from stem cells in the peripheral blood (blood outgrowth endothelial cells) and PBMCs from the same donor can be used to create an autologous coculture bioassay that responds by releasing a plethora of cytokines to authentic TGN1412 but only modestly to Campath and not to control antibodies such as Herceptin, Avastin, and Arzerra. This assay performed better than the traditional mixed donor assay in terms of cytokine release to TGN1412 and, thus, we suggest provides significant advancement and a definitive system by which biologics can be tested and paves the way for personalized medicine.


Assuntos
Produtos Biológicos/farmacologia , Citocinas/metabolismo , Células Endoteliais/efeitos dos fármacos , Leucócitos Mononucleares/efeitos dos fármacos , Alemtuzumab , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados/farmacologia , Bevacizumab , Bioensaio/métodos , Proliferação de Células/efeitos dos fármacos , Técnicas de Cocultura , Meios de Cultura/farmacologia , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Ensaio de Imunoadsorção Enzimática , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Interleucina-2/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/metabolismo , Reprodutibilidade dos Testes , Soro/química , Trastuzumab , Fator de Necrose Tumoral alfa/metabolismo
6.
FASEB J ; 28(1): 373-81, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24072781

RESUMO

Gene therapy is a powerful approach to treat disease locally. However, if the therapeutic target is intracellular, the therapeutic will be effective only in the cells where the therapeutic gene is delivered. We have engineered a fusion protein containing an intracellular inhibitor of the transcription factor NF-κB pathway that can be effectively secreted from producing cells. This fusion protein is cleaved extracellularly by metalloproteinases allowing release of a protein transduction domain (PTD) linked to the NF-κB inhibitor for translocation into neighboring cells. We show that engineered molecules can be efficiently secreted (>80%); are cleaved with matrix metalloprotease-1; inhibit NF-κB driven transcription in a biological assay with a human reporter cell line; and display significant inhibition in mouse paw inflammation models when delivered by lentivirus or secreting cells. No inhibition of NF-κB transcription or therapeutic effect was seen using molecules devoid of the PTD and NF-κB inhibitory domains. By creating a fusion protein with an endogenous secretion partner, we demonstrate a novel approach to efficiently secrete PTD-containing protein domains, overcoming previous limitations, and allowing for potent paracrine effects.


Assuntos
NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Linhagem Celular , Terapia Genética/métodos , Humanos , Metaloproteinase 1 da Matriz/genética , Metaloproteinase 1 da Matriz/metabolismo , Proteínas Recombinantes de Fusão/genética
7.
Ann Rheum Dis ; 73(9): 1728-36, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23813971

RESUMO

BACKGROUND: Latent cytokines are engineered by fusing the latency associated peptide (LAP) derived from transforming growth factor-ß (TGF-ß) with the therapeutic cytokine, in this case interferon-ß (IFN-ß), via an inflammation-specific matrix metalloproteinase (MMP) cleavage site. OBJECTIVES: To demonstrate latency and specific delivery in vivo and to compare therapeutic efficacy of aggrecanase-mediated release of latent IFN-ß in arthritic joints to the original MMP-specific release. METHODS: Recombinant fusion proteins with MMP, aggrecanase or devoid of cleavage site were expressed in CHO cells, purified and characterised in vitro by Western blotting and anti-viral protection assays. Therapeutic efficacy and half-life were assessed in vivo using the mouse collagen-induced arthritis model (CIA) of rheumatoid arthritis and a model of acute paw inflammation, respectively. Transgenic mice with an IFN-regulated luciferase gene were used to assess latency in vivo and targeted delivery to sites of disease. RESULTS: Efficient localised delivery of IFN-ß to inflamed paws, with low levels of systemic delivery, was demonstrated in transgenic mice using latent IFN-ß. Engineering of latent IFN-ß with an aggrecanase-sensitive cleavage site resulted in efficient cleavage by ADAMTS-4, ADAMTS-5 and synovial fluid from arthritic patients, with an extended half-life similar to the MMP-specific molecule and greater therapeutic efficacy in the CIA model. CONCLUSIONS: Latent cytokines require cleavage in vivo for therapeutic efficacy, and they are delivered in a dose dependent fashion only to arthritic joints. The aggrecanase-specific cleavage site is a viable alternative to the MMP cleavage site for the targeting of latent cytokines to arthritic joints.


Assuntos
Antirreumáticos/administração & dosagem , Artrite Experimental/tratamento farmacológico , Artrite Reumatoide/tratamento farmacológico , Interferon beta/administração & dosagem , Animais , Antirreumáticos/farmacocinética , Antirreumáticos/uso terapêutico , Células CHO , Cricetulus , Citocinas , Relação Dose-Resposta a Droga , Sistemas de Liberação de Medicamentos/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Endopeptidases , Meia-Vida , Interferon beta/farmacocinética , Interferon beta/uso terapêutico , Metaloproteinases da Matriz , Camundongos , Camundongos Transgênicos , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/uso terapêutico
8.
J Vis Exp ; (199)2023 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-37782084

RESUMO

New immunostimulatory antibody drugs designed to either directly stimulate specific immune cells or indirectly enhance the immune response by blocking or activating an endogenous regulator of the immune system have the potential to cause serious immune-related adverse events such as cytokine release syndrome (CRS). It is, therefore crucial to assess the safety profile of such drugs with a combination of in vivo and in vitro experiments before first-in-human dose administration. Cytokine release assays (CRAs), where the proposed antibody therapeutic is co-cultured with human immune cells (such as peripheral blood mononuclear cells (PBMCs), whole blood, or otherwise) and the amount of inflammatory cytokine produced is measured, are critical for hazard identification. However, different labs using different control antibodies can threaten the harmonization of CRAs, and clinically relevant controls (such as TGN1412) can be difficult to source, which can lead to less accurate or reliable results or data which are difficult to compare between laboratories. The inclusion of positive and negative controls in a CRA can ensure the accuracy and reliability of the results. The National Institute for Biological Standards and Control (NIBSC) has produced a panel of lyophilized antibody controls intended for use in various CRA platforms to harmonize results across various laboratories and assay methods. A set of three different positive control antibodies include anti-CD52, anti-CD3, and anti-CD28 superagonist (SA), which are known to induce dose-dependent CRS in patients. Each antibody is provided with an isotype-matched negative control antibody. This panel of reference reagents has previously been shown to have good inter-lab reproducibility and are suitable controls to increase the confidence and robustness of safety data from a variety of CRA platforms.


Assuntos
Anticorpos Monoclonais , Leucócitos Mononucleares , Humanos , Anticorpos Monoclonais/farmacologia , Reprodutibilidade dos Testes , Citocinas , Antígenos CD28
9.
Ann Rheum Dis ; 71(1): 143-9, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21998117

RESUMO

OBJECTIVE: To facilitate the targeting to inflammation sites of small anti-inflammatory peptides, with short half-lives, by fusion with the latency-associated peptide (LAP) of transforming growth factor ß1 through a cleavable matrix metalloproteinase (MMP) linker. This design improves efficacy, overcoming the limitations to their clinical use. METHODS: We generated latent forms of vasoactive intestinal peptide (VIP), α-melanocyte-stimulating hormone (MSH) and γ(3)MSH by fusion to LAP through an MMP cleavage site using recombinant DNA technology. The biological activities of these latent therapeutics were studied in vivo using monosodium urate (MSU)-induced peritonitis and collagen-induced arthritis (CIA) models. We assessed gene therapy and purified protein therapy. RESULTS: The recruitment of the polymorphonuclear cells induced by MSU injection into mouse peritoneal cavity was reduced by 35% with γ(3)MSH (1 nmol), whereas administration of a much lower dose of purified latent LAP-MMP-γ(3)MSH (0.03 nmol) attenuated leucocyte influx by 50%. Intramuscular gene delivery of plasmids coding LAP-MMP-VIP and LAP-MMP-αMSH at disease onset reduced the development of CIA compared with LAP-MMP, which does not contain any therapeutic moiety. Histological analysis confirmed a significantly lower degree of inflammation, bone and cartilage erosion in groups treated with LAP-MMP-VIP or LAP-MMP-αMSH. Antibody titres to collagen type II and inflammatory cytokine production were also reduced in these two groups. CONCLUSION: Incorporation of small anti-inflammatory peptides within the LAP shell and delivered as recombinant protein or through gene therapy can control inflammatory and arthritic disease. This platform delivery can be developed to control human arthritides and other autoimmune diseases.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Artrite Experimental/terapia , Hormônios Estimuladores de Melanócitos/uso terapêutico , Fragmentos de Peptídeos/uso terapêutico , Peritonite/terapia , Proteínas Recombinantes de Fusão/uso terapêutico , Fator de Crescimento Transformador beta/uso terapêutico , Peptídeo Intestinal Vasoativo/uso terapêutico , Animais , Anti-Inflamatórios não Esteroides/farmacocinética , Artrite Experimental/tratamento farmacológico , Artrite Experimental/metabolismo , Artrite Experimental/patologia , Citocinas/sangue , Sistemas de Liberação de Medicamentos , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos/métodos , Terapia Genética/métodos , Meia-Vida , Masculino , Hormônios Estimuladores de Melanócitos/genética , Hormônios Estimuladores de Melanócitos/farmacocinética , Camundongos , Camundongos Endogâmicos DBA , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/farmacocinética , Peritonite/tratamento farmacológico , Peritonite/metabolismo , Proteínas Recombinantes de Fusão/farmacocinética , Distribuição Tecidual , Resultado do Tratamento , Peptídeo Intestinal Vasoativo/genética , Peptídeo Intestinal Vasoativo/farmacocinética
10.
Sci Rep ; 12(1): 18694, 2022 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-36333445

RESUMO

SARS-CoV-2 exhibits a diverse host species range with variable outcomes, enabling differential host susceptibility studies to assess suitability for pre-clinical countermeasure and pathogenesis studies. Baseline virological, molecular and pathological outcomes were determined among multiple species-one Old World non-human primate (NHP) species (cynomolgus macaques), two New World NHP species (red-bellied tamarins; common marmosets) and Syrian hamsters-following single-dose, atraumatic intranasal administration of SARS-CoV-2/Victoria-01. After serial sacrifice 2, 10 and 28-days post-infection (dpi), hamsters and cynomolgus macaques displayed differential virus biodistribution across respiratory, gastrointestinal and cardiovascular systems. Uniquely, New World tamarins, unlike marmosets, exhibited high levels of acute upper airway infection, infectious virus recovery associated with mild lung pathology representing a host previously unrecognized as susceptible to SARS-CoV-2. Across all species, lung pathology was identified post-clearance of virus shedding (antigen/RNA), with an association of virus particles within replication organelles in lung sections analysed by electron microscopy. Disrupted cell ultrastructure and lung architecture, including abnormal morphology of mitochondria 10-28 dpi, represented on-going pathophysiological consequences of SARS-CoV-2 in predominantly asymptomatic hosts. Infection kinetics and host pathology comparators using standardized methodologies enables model selection to bridge differential outcomes within upper and lower respiratory tracts and elucidate longer-term consequences of asymptomatic SARS-CoV-2 infection.


Assuntos
COVID-19 , SARS-CoV-2 , Cricetinae , Animais , Distribuição Tecidual , Administração Intranasal , Modelos Animais de Doenças , Pulmão/patologia , Mesocricetus , Macaca fascicularis
11.
Viruses ; 13(8)2021 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-34452392

RESUMO

Lentiviral vectors have played a critical role in the emergence of gene-modified cell therapies, specifically T cell therapies. Tisagenlecleucel (Kymriah), axicabtagene ciloleucel (Yescarta) and most recently brexucabtagene autoleucel (Tecartus) are examples of T cell therapies which are now commercially available for distribution after successfully obtaining EMA and FDA approval for the treatment of blood cancers. All three therapies rely on retroviral vectors to transduce the therapeutic chimeric antigen receptor (CAR) into T lymphocytes. Although these innovations represent promising new therapeutic avenues, major obstacles remain in making them readily available tools for medical care. This article reviews the biological principles as well as the bioprocessing of lentiviral (LV) vectors and adoptive T cell therapy. Clinical and engineering successes, shortcomings and future opportunities are also discussed. The development of Good Manufacturing Practice (GMP)-compliant instruments, technologies and protocols will play an essential role in the development of LV-engineered T cell therapies.


Assuntos
Engenharia Celular/métodos , Vetores Genéticos , Lentivirus/genética , Receptores de Antígenos Quiméricos/genética , Linfócitos T/imunologia , Ensaios Clínicos como Assunto , Humanos , Imunoterapia Adotiva/métodos , Neoplasias/terapia , Receptores de Antígenos Quiméricos/imunologia
12.
Arterioscler Thromb Vasc Biol ; 29(11): 1757-63, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19661485

RESUMO

OBJECTIVE: The proinflammatory phenotype induced by low laminar shear stress (LSS) is implicated in atherogenesis. The kinin B1 receptor (B1R), known to be induced by inflammatory stimuli, exerts many proinflammatory effects including vasodilatation and leukocyte recruitment. We investigated whether low LSS is a stimulus for endothelial B1R expression and function. METHODS AND RESULTS: Human and mouse atherosclerotic plaques expressed high level of B1R mRNA and protein. In addition, B1R expression was upregulated in the aortic arch (low LSS region) of ApoE(-/-) mice fed a high-fat diet compared to vascular regions of high LSS and animals fed normal chow. Of interest, a greater expression of B1R was noticed in endothelial cells from regions of low LSS in aortic arch of ApoE(-/-) mice. B1R was also upregulated in human umbilical vein endothelial cells (HUVECs) exposed to low LSS (0 to 2 dyn/cm(2)) compared to physiological LSS (6 to 10 dyn/cm(2)): an effect similarly evident in murine vascular tissue perfused ex vivo. Functionally, B1R activation increased prostaglandin and CXCL5 expression in cells exposed to low, but not physiological, LSS. IL-1beta and ox-LDL induced B1R expression and function in HUVECs, a response substantially enhanced under low LSS conditions and inhibited by blockade of NFkappaB activation. CONCLUSIONS: Herein, we show that LSS is a major determinant of functional B1R expression in endothelium. Furthermore, whereas physiological high LSS is a powerful repressor of this inflammatory receptor, low LSS occurring [corrected] at sites of atheroma is associated with substantial upregulation, identifying this receptor as a potential therapeutic target.


Assuntos
Aterosclerose/genética , Aterosclerose/metabolismo , Endotélio Vascular/metabolismo , Receptor B1 da Bradicinina/metabolismo , Estresse Fisiológico/fisiologia , Animais , Aorta Torácica/metabolismo , Apolipoproteínas E/metabolismo , Sítios de Ligação , Western Blotting , Células Cultivadas , Endotélio Vascular/citologia , Regulação da Expressão Gênica , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Resistência ao Cisalhamento/fisiologia , Transdução de Sinais , Estresse Mecânico , Veias Umbilicais/citologia , Veias Umbilicais/metabolismo
13.
Artigo em Inglês | MEDLINE | ID: mdl-34095644

RESUMO

Accurate determination of cellular subsets that secrete particular cytokine(s) is a significant parameter for functional characterization of an immunological response. The present study was conducted to develop a method for simultaneous measurement of intracellular cytokine positive CD4 and CD8 positive T lymphocytes in a single tube, with a no-wash protocol. We report here the development of a simplified, rapid procedure for precise enumeration of cytokine positive T lymphocytes using BD Trucount tubes. This single step protocol for accurate enumeration of cytokine positive T lymphocytes, will allow for better characterization of immune cell phenotype and function.

14.
Cytokine X ; 2(4): 100042, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33458650

RESUMO

Immunomodulatory therapeutics such as monoclonal antibodies (mAb) carry an inherent risk of undesired immune reactions. One such risk is cytokine release syndrome (CRS), a rapid systemic inflammatory response characterized by the secretion of pro-inflammatory cytokines from immune cells. It is crucial for patient safety to correctly identify potential risk of CRS prior to first-in-human dose administration. For this purpose, a variety of in vitro cytokine release assays (CRA) are routinely used as part of the preclinical safety assessment of novel therapeutic mAbs. One of the challenges for the development and comparison of CRA performance is the lack of availability of standard positive and negative control mAbs for use in assay qualification. To address this issue, the National Institute for Biological Standards and Control (NIBSC) developed a reference panel of lyophilised mAbs known to induce CRS in the clinic: human anti-CD52, mouse anti-CD3 and human superagonistic (SA) anti-CD28 mAb manufactured according to the respective published sequences of Campath-1H® (alemtuzumab, IgG1) , Orthoclone OKT-3® (muromonab, IgG2a) and TGN1412 (theralizumab, IgG4), as well as three isotype matched negative controls (human IgG1, mouse IgG2a and human IgG4, respectively). The relative capacity of these control mAbs to stimulate the release of IFN-γ, IL-2, TNF-α and IL-6 in vitro was evaluated in eleven laboratories in an international collaborative study mediated through the HESI Immuno-safety Technical Committee Cytokine Release Assay Working Group. Participants tested the NIBSC mAbs in a variety of CRA platforms established at each institution. This paper presents the results from the centralised cytokine quantification on all the plasma/supernatants corresponding to the stimulation of immune cells in the different CRA platforms by a single concentration of each mAb. Each positive control mAb induced significant cytokine release in most of the tested CRA platforms. There was a high inter-laboratory variability in the levels of cytokines produced, but similar patterns of response were observed across laboratories that replicated the cytokine release patterns previously published for the respective clinical therapeutic mAbs. Therefore, the positive and negative mAbs are suitable as a reference panel for the qualification and validation of CRAs, comparison of different CRA platforms (e.g. solid vs aqueous phase), and intra- and inter-laboratory comparison of CRA performance. Thus, the use of this panel of positive and negative control mAbs will increase the confidence in the robustness of a CRA platform to identify a potential CRS risk for novel immunomodulatory therapeutic candidates.

15.
Artigo em Inglês | MEDLINE | ID: mdl-31993416

RESUMO

The symptomatic irreproducibility of data in biomedicine and biotechnology prompts the need for higher order measurements of cells in their native and near-native environments. Such measurements may support the adoption of new technologies as well as the development of research programs across different sectors including healthcare and clinic, environmental control and national security. With an increasing demand for reliable cell-based products and services, cellular metrology is poised to help address current and emerging measurement challenges faced by end-users. However, metrological foundations in cell analysis remain sparse and significant advances are necessary to keep pace with the needs of modern medicine and industry. Herein we discuss a role of metrology in cell and cell-related R&D activities to underpin growing international measurement capabilities. Relevant measurands are outlined and the lack of reference methods and materials, particularly those based on functional cell responses in native environments, is highlighted. The status quo and current challenges in cellular measurements are discussed in the light of metrological traceability in cell analysis and applications (e.g., a functional cell count). An emphasis is made on the consistency of measurement results independent of the analytical platform used, high confidence in data quality vs. quantity, scale of measurements and issues of building infrastructure for end-users.

16.
Cytometry B Clin Cytom ; 96(6): 508-513, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30790450

RESUMO

BACKGROUND: Over 2,000 people a year in the United Kingdom need a bone marrow or blood stem cell transplant. It is important to accurately quantify the hematopoietic stem cells to predict whether the transplant will be successful in replenishing the immune system. However, they are present at low frequency, which complicates accurate quantification. The current gold standard method is single-platform flow cytometry using internal reference counting beads to determine the concentration of CD34 cells. However, volumetric flow cytometers have the ability to measure the acquisition volume, which removes the need for reference beads for calculation of cell concentrations. METHOD: In this study, we compared both methods for calculating CD34 cell concentrations in volumetric cytometers, using either the volume reading or the number of reference beads for calculation. In addition, the uncertainty of measurement for each method was estimated. RESULTS: The results show that both methods have similar uncertainties of measurement. Regression analysis showed low to no statistical difference in CD34 cell concentrations obtained with each method. CONCLUSIONS: Overall, this study suggests that the volumetric method is a valid approach but that the adoption of this technology may be hindered without some form of external calibration of volume readings to increase confidence in the measurement. © 2019 The Authors. Cytometry Part B: Clinical Cytometry published by Wiley Periodicals, Inc. on behalf of International Clinical Cytometry Society.


Assuntos
Antígenos CD34/análise , Citometria de Fluxo , Células-Tronco Hematopoéticas/citologia , Contagem de Células , Humanos , Análise de Regressão
17.
Nat Biotechnol ; 21(11): 1314-20, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14528315

RESUMO

To increase the half-life of a cytokine and target its activation specifically to disease sites, we have engineered a latent cytokine using the latency-associated protein (LAP) of transforming growth factor-beta 1 (TGF-beta 1) fused via a matrix metalloproteinase (MMP) cleavage site to interferon (IFN)-beta at either its N or C terminus. The configuration LAP-MMP-IFN-beta resembles native TGF-beta and lacks biological activity until cleaved by MMPs, whereas the configuration IFN-beta-MMP-LAP is active. LAP provides for a disulfide-linked shell hindering interaction of the cytokine with its cellular receptors, conferring a very long half-life of 55 h in vivo. Mutations of the disulfide bonds in LAP abolish this latency. Samples of cerebrospinal fluid (CSF) or synovial fluid from patients with inflammatory diseases specifically activate the latent cytokine, whereas serum samples do not. Intramuscular injection in arthritic mice of plasmid DNA encoding these constructs demonstrated a greater therapeutic effect of the latent as compared to the active forms.


Assuntos
Quimiocinas/imunologia , Quimiocinas/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Marcação de Genes/métodos , Receptores de Quimiocinas/imunologia , Receptores de Quimiocinas/metabolismo , Animais , Células Cultivadas , Quimiocinas/administração & dosagem , Quimiocinas/genética , Retículo Endoplasmático/imunologia , Retículo Endoplasmático/metabolismo , Humanos , Camundongos , Osteíte/imunologia , Osteíte/metabolismo , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo
18.
Trends Mol Med ; 10(6): 269-74, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15177191

RESUMO

Cytokines are mediators of cell communication. Their therapeutic use requires frequent high doses to achieve effective local biological levels. However, the clinical use of some cytokines is limited because of their pleiotropism, which can result in unwanted side effects. Here, we review novel protein engineering technologies that overcome these limitations and enable the targeting of cytokines to specific sites. One such technology uses antibody-based recognition to direct the cytokine to a particular tissue, and another creates encapsulated latent cytokines that are released only at the site of disease. The latter method requires the overexpression of matrix-metalloproteinases, thereby exploiting the severity of the pathological process to regulate drug delivery. Because these technologies are based on the expression of fusion proteins, their application can be extended to other biologicals and can be delivered by gene therapy.


Assuntos
Portadores de Fármacos , Sistemas de Liberação de Medicamentos , Engenharia de Proteínas , Proteínas Recombinantes de Fusão , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/uso terapêutico , Arteriosclerose/fisiopatologia , Arteriosclerose/terapia , Doenças Autoimunes/fisiopatologia , Doenças Autoimunes/terapia , Comunicação Celular/fisiologia , Doenças Transmissíveis/fisiopatologia , Doenças Transmissíveis/terapia , Citocinas/genética , Citocinas/uso terapêutico , Sistemas de Liberação de Medicamentos/métodos , Sistemas de Liberação de Medicamentos/tendências , Terapia Genética/métodos , Humanos , Metaloproteases/metabolismo , Neoplasias/fisiopatologia , Neoplasias/terapia , Proteínas Recombinantes de Fusão/genética
19.
Protein Eng Des Sel ; 17(12): 829-35, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15708865

RESUMO

We have engineered a latent mouse interferon beta (mIFNbeta) using the latency associated peptide (LAP) of transforming growth factor beta1 (TGF-beta1) to protect the cytokine and avoid its interaction with its receptors. This approach improves the pharmacokinetic properties and reduces the pleiotropic effects limiting the current therapeutic use of cytokines. IFNbeta was fused to the LAP using two flexible linkers flanking a matrix metalloproteinase (MMP) cleavage site for the specific release of IFNbeta at disease sites. In order to improve the hydrolysis rate of the cleavage site, 15 different cleavable linkers were introduced in the LAP-mIFNbeta construct. The kinetic parameters relative to the linker cleavage by MMP-1 and MMP-3 were measured by an ELISA method. Among the modifications done, one of the constructs bearing the activation site of pro-MMPs was the best substrate for both enzymes. The introduction of a hydrophilic sequence derived from the furin cleavage site of the anthrax toxin protective antigen increased the sensitivity to MMP-3 to up to 29-fold. These data suggest that this strategy could be useful for improving the effectiveness of the delivery and targeting of protein therapeutics.


Assuntos
Citocinas/biossíntese , Metaloproteinase 1 da Matriz/metabolismo , Metaloproteinase 3 da Matriz/metabolismo , Engenharia de Proteínas/métodos , Animais , Antígenos de Bactérias/química , Toxinas Bacterianas/química , Sítios de Ligação , Domínio Catalítico , Clonagem Molecular , Citocinas/metabolismo , Primers do DNA/química , Ensaio de Imunoadsorção Enzimática , Humanos , Hidrólise , Interferon beta/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/química , Cinética , Metaloproteinases da Matriz/metabolismo , Camundongos , Peptídeos/química , Plasmídeos/metabolismo , Ligação Proteica , Sensibilidade e Especificidade , Fatores de Tempo , Transfecção
20.
Expert Opin Drug Deliv ; 11(1): 101-10, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24294995

RESUMO

INTRODUCTION: The use of cytokines as therapeutic agents is important, given their potent biological effects. However, this very potency, coupled with the pleiotropic nature and short half-life of these molecules, has limited their therapeutic use. Strategies to increase the half-life and to decrease toxicity are necessary to allow effective treatment with these molecules. AREAS COVERED: A number of strategies are used to overcome the natural limitations of cytokines, including PEGylation, encapsulation in liposomes, fusion to targeting peptides or antibodies and latent cytokines. Latent cytokines are engineered using the latency-associated peptide of transforming growth factor-ß to produce therapeutic cytokines/peptides that are released only at the site of disease by cleavage with disease-induced matrix metalloproteinases. The principles underlying the latent cytokine technology are described and are compared to other methods of cytokine delivery. The potential of this technology for developing novel therapeutic strategies for the treatment of diseases with an inflammatory-mediated component is discussed. EXPERT OPINION: Methods of therapeutic cytokine delivery are addressed. The latent cytokine technology holds significant advantages over other methods of drug delivery by providing simultaneously increased half-life and localised drug delivery without systemic effects. Cytokines that failed clinical trials should be reassessed using this delivery system.


Assuntos
Citocinas/administração & dosagem , Sistemas de Liberação de Medicamentos , Peptídeos/administração & dosagem , Precursores de Proteínas/administração & dosagem , Fator de Crescimento Transformador beta/administração & dosagem , Animais , Citocinas/química , Humanos , Inflamação/tratamento farmacológico , Peptídeos/química , Precursores de Proteínas/química , Fator de Crescimento Transformador beta/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA