Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Physiother Theory Pract ; : 1-14, 2023 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-37403743

RESUMO

INTRODUCTION: Cadaveric dissection shapes the ways in which healthcare students understand the human body and the attitudes, identities and behaviors they exhibit as health professionals. There is however a paucity of related research with physiotherapy (PT) students. PURPOSE: The purpose of this interpretivist study was to investigate PT students' conceptions of the human body in relation to experiences with human cadavers in anatomy education. METHODS: Ten semi-structured interviews were conducted with PT students along with four optional written reflections completed. Data was thematically analyzed. RESULTS: Students engaged in a continuous process of habituation involving oscillation between "humanization" and "dehumanization" of cadavers in the anatomy lab. We describe the contextual mediators that shaped the process, the multi-sensory and emotional experience of the students, and the "interruptions" that contributed to the variability in their conceptions over time and contexts. Students ultimately habituated toward dehumanization which had multiple effects on learning and professionalization. CONCLUSION: Study findings highlight the complexity of PT students' experiences and learning within the cadaver lab outside of the formal goals of anatomy education. We discuss the implications for anatomy curricula, including the potential advantages of incorporating a biopsychosocial approach.

2.
Thyroid ; 33(6): 732-742, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37016815

RESUMO

Background: Treatment of Graves' hyperthyroidism (GH) and Graves' orbitopathy (GO) is far from adequate, and hence, new substances that specifically target the autoantigens in GH/GO are warranted. This study determined the preclinical in vitro efficacy of SYD5115, a novel low-molecular-weight compound that inhibits the thyrotropin receptor (TSH-R). Methods: The TSH-R inhibiting capability of SYD5115 was tested through stimulation of wild-type and chimeric TSH-R expressed in Chinese hamster ovary (CHO) cells using two functional (stimulatory and blocking) cell-based TSH-R-Ab bioassays. TSH-R expressing human orbital fibroblasts, collected from GH+GO patients (GOF), were stimulated with the monoclonal antibody M22 or with stimulatory TSH-R-Ab (TSAb)-positive sera with cyclic adenosine monophosphate (cAMP) or hyaluronic acid (HA) release as readouts. The effect of SYD5115 on the viability of GOF was tested in 4,5-dimethylthiazol-2-yl-2,5-diphenyltetrazolium bromide and scratch cell growth assays. Results: SYD5115 significantly and dose dependently inhibited the TSH-R activation through M22 or TSAb-positive sera in all performed bioassays. Inhibition showed similar levels in the TSAb reporter bioassay and in the cAMP assay with GOF. The % inhibition and compound concentration showed a sigmoidal relationship, with all seven TSAb-positive sera markedly inhibited by SYD5115. An SYD5115 dose-dependent inhibition of M22 (10 ng/mL, 6 hours)-stimulated HA and/or cAMP-release from GOF was observed. Strong SYD5115-induced inhibitions of M22-stimulated cAMP production in GOF were registered with SYD5115 concentrations of 1 (p = 0.0029), 10 (p < 0.0001), 100 (p < 0.0001), 1,000 (p < 0.0001), and 10,000 (p < 0.0001) nM, respectively. SYD5115-induced inhibition of M22-stimulated HA production was noted with SYD5115 concentrations of 100 (p = 0.0392), 1000 (p = 0.0431), and 10,000 (p = 0.0245) nM, respectively. The inhibitory activity of SYD5115 was confirmed in a human osteosarcoma U2OS cell line stably expressing human TSH-R with cAMP as readout. SYD5115 induced 100% inhibition of the M22-induced cAMP levels with a potency of 193 nM. Compared with control, SYD5115 did neither impact the growth nor the migration of cultivated GOF. In addition, SYD5115 did not alter the viability of GOF. Conclusions: SYD5115 blocked M22- and TSAb-induced TSH-R activity with a nanomolar potency in TSH-R-overexpressed CHO cells as well as primary GOF, which demonstrates the ability of this small molecule to block TSH-R overactivity.


Assuntos
Oftalmopatia de Graves , Receptores da Tireotropina , Cricetinae , Animais , Humanos , Oftalmopatia de Graves/tratamento farmacológico , Cricetulus , Células CHO , Imunoglobulinas Estimuladoras da Glândula Tireoide , Tireotropina/metabolismo , Autoanticorpos
3.
Mol Cancer Ther ; 22(6): 765-777, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37042205

RESUMO

MET, the cell-surface receptor for the hepatocyte growth factor/scatter factor, which is widely overexpressed in various solid cancer types, is an attractive target for the development of antibody-based therapeutics. BYON3521 is a novel site-specifically conjugated duocarmycin-based antibody-drug conjugate (ADC), comprising a humanized cysteine-engineered IgG1 monoclonal antibody with low pmol/L binding affinity towards both human and cynomolgus MET. In vitro studies showed that BYON3521 internalizes efficiently upon MET binding and induces both target- and bystander-mediated cell killing. BYON3521 showed good potency and full efficacy in MET-amplified and high MET-expressing cancer cell lines; in moderate and low MET-expressing cancer cell lines good potencies and partial efficacy were observed. In mouse xenograft models, BYON3521 showed significant antitumor activity upon single-dose administration in multiple non-MET-amplified tumor types with low, moderate, and high MET expression, including complete tumor remissions in models with moderate MET expression. In the repeat-dose Good Laboratory Practice (GLP) safety assessment in cynomolgus monkeys, BYON3521 was well tolerated and based on the observed toxicities and their reversibility, the highest non-severely toxic dose was set at 15 mg/kg. A human pharmacokinetics (PK) model was derived from the PK data from the cynomolgus safety assessments, and the minimal efficacious dose in humans is estimated to be in the range of 3 to 4 mg/kg. In all, our nonclinical data suggests that BYON3521 is a safe ADC with potential for clinical benefit in patients. A first-in-human dose-escalation study is currently ongoing to determine the maximum tolerated dose and recommended dose for expansion (NCT05323045).


Assuntos
Anticorpos Monoclonais , Imunoconjugados , Animais , Humanos , Camundongos , Anticorpos Monoclonais Humanizados , Linhagem Celular Tumoral , Imunoglobulina G , Ensaios Antitumorais Modelo de Xenoenxerto
4.
J Immunother Cancer ; 11(4)2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-37068796

RESUMO

BACKGROUND: Preclinical studies have firmly established the CD47-signal-regulatory protein (SIRP)α axis as a myeloid immune checkpoint in cancer, and this is corroborated by available evidence from the first clinical studies with CD47 blockers. However, CD47 is ubiquitously expressed and mediates functional interactions with other ligands as well, and therefore targeting of the primarily myeloid cell-restricted inhibitory immunoreceptor SIRPα may represent a better strategy. METHOD: We generated BYON4228, a novel SIRPα-directed antibody. An extensive preclinical characterization was performed, including direct comparisons to previously reported anti-SIRPα antibodies. RESULTS: BYON4228 is an antibody directed against SIRPα that recognizes both allelic variants of SIRPα in the human population, thereby maximizing its potential clinical applicability. Notably, BYON4228 does not recognize the closely related T-cell expressed SIRPγ that mediates interactions with CD47 as well, which are known to be instrumental in T-cell extravasation and activation. BYON4228 binds to the N-terminal Ig-like domain of SIRPα and its epitope largely overlaps with the CD47-binding site. BYON4228 blocks binding of CD47 to SIRPα and inhibits signaling through the CD47-SIRPα axis. Functional studies show that BYON4228 potentiates macrophage-mediated and neutrophil-mediated killing of hematologic and solid cancer cells in vitro in the presence of a variety of tumor-targeting antibodies, including trastuzumab, rituximab, daratumumab and cetuximab. The silenced Fc region of BYON4228 precludes immune cell-mediated elimination of SIRPα-positive myeloid cells, implying anticipated preservation of myeloid immune effector cells in patients. The unique profile of BYON4228 clearly distinguishes it from previously reported antibodies representative of agents in clinical development, which either lack recognition of one of the two SIRPα polymorphic variants (HEFLB), or cross-react with SIRPγ and inhibit CD47-SIRPγ interactions (SIRPAB-11-K322A, 1H9), and/or have functional Fc regions thereby displaying myeloid cell depletion activity (SIRPAB-11-K322A). In vivo, BYON4228 increases the antitumor activity of rituximab in a B-cell Raji xenograft model in human SIRPαBIT transgenic mice. Finally, BYON4228 shows a favorable safety profile in cynomolgus monkeys. CONCLUSIONS: Collectively, this defines BYON4228 as a preclinically highly differentiating pan-allelic SIRPα antibody without T-cell SIRPγ recognition that promotes the destruction of antibody-opsonized cancer cells. Clinical studies are planned to start in 2023.


Assuntos
Antígeno CD47 , Neoplasias , Camundongos , Animais , Humanos , Linfócitos T/metabolismo , Rituximab , Macrófagos , Neoplasias/tratamento farmacológico , Anticorpos Antineoplásicos
5.
Bioorg Med Chem ; 84: 117258, 2023 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-37001244

RESUMO

The thyrotropin receptor (TSH-R) regulates the thyroid gland and is normally activated by thyrotropin. In patients with Graves' disease, TSH-R is also stimulated by stimulatory TSH-R autoantibodies leading to hyperthyroidism. In this paper, we describe the discovery of SYD5115 (67), a novel small molecule TSH-R antagonist with nanomolar potency. SYD5115 also blocks stimulating antibody induced synthesis of the thyroid hormone thyroxine (T4) in vivo, after a single oral dose. During optimization, several issues had to be addressed such as the low metabolic stability and the potential mutagenicity of our first series of compounds.


Assuntos
Doença de Graves , Receptores da Tireotropina , Humanos , Autoanticorpos , Doença de Graves/tratamento farmacológico , Receptores Acoplados a Proteínas G , Receptores da Tireotropina/antagonistas & inibidores , Tireotropina/metabolismo
6.
J Appl Soc Sci (Boulder) ; 16(2): 442-458, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35873708

RESUMO

Background and aim: U.S. state governments have the responsibility to regulate and license behavioral healthcare interventions, such as for addiction and mental illness, with increasing emphasis on implementing evidence-based programs (EBPs). A serious obstacle to this is lack of clarity or agreement about what constitutes "evidence-based." The study's purpose was to determine the extent to which and in what contexts web-based Evidence-based Program Registries (EBPRs) are referenced in state government statutes and regulations ("mandates") concerning behavioral healthcare. Examples are: What Works Clearinghouse; National Register of Evidence-based Programs and Practices; Cochrane Database of Systematic Reviews. Methods: The study employed the Westlaw Legal Research Database to search for 30 known EBPR websites relevant to behavioral healthcare within the statutes and regulations of all 50 states. Results: There was low prevalence of EBPR references in state statutes and regulations pertaining to behavioral healthcare; 20 states had a total of 33 mandates that referenced an EBPR. These mandates usually do not rely on an EBPR as the sole acceptable source for classifying a program or practice as "evidence-based." Instead, EBPRs were named in conjunction with internal state or external sources of information about putative program effectiveness, which may be less valid than EBPRs, to determine what is "evidence-based." Conclusion: Greater awareness of scientifically - based EBPRs and greater understanding of their advantages need to be fostered among state legislators and regulators charged with making policy to increase or improve the use of evidence-based programs and practices in behavioral healthcare in the U.S.

7.
Eval Health Prof ; 45(4): 397-410, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35446692

RESUMO

Evidence-based program registries (EBPRs) are web-based compilations of behavioral healthcare programs/interventions that rely on research-based criteria to rate program efficacy or effectiveness for support of programmatic decision-making. The objective was to determine the extent to which behavioral health decision-makers access EBPRs and to understand whether and exactly how they use the information obtained from EPBRs. Single State Authorities (SSAs) and service provider agencies in the areas of behavioral health and child welfare were recruited nationally. Senior staff (n = 375) responsible for the selection and implementation of programs and/or policies were interviewed by telephone concerning their visits (if any) to 28 relevant EBPRs, the types of information they were seeking, whether they found it, and how they may have used that information to effect changes in their organizations. At least one EBPR was visited by 80% of the respondents, with a median of three different registers being visited. Most visitors (55%) found all the information they were seeking; those who did not desired more guidance or tools for individual program implementation or were unable to locate the program or practice that they were seeking. Most visitors (65%) related using the information obtained to make changes in their organizations, in particular to select, start or change a program, or to support the adoption or improvement of evidence-based clinical practices. EBPRs were shown to be important resources for dissemination of research-based program effectiveness data, leading to increased use of evidence-based practices in the field, but the study also identified needs for greater awareness of EBPRs generally and for more attention to implementation of specific recommended programs and practices.


Assuntos
Atenção à Saúde , Prática Clínica Baseada em Evidências , Criança , Humanos , Avaliação de Programas e Projetos de Saúde , Sistema de Registros , Proteção da Criança
8.
Eval Program Plann ; 85: 101906, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33567376

RESUMO

PURPOSE: Evidence-based program registers (EBPRs) are important tools for facilitating the use of evidence-based practices or programs (EBPs) by state statutory agencies responsible for behavioral healthcare, broadly defined as substance misuse, mental health, HIV/AIDS prevention, child welfare, and offender rehabilitation. There are currently no data on the purposes for which such state agencies reference EBPRs on their official websites. METHOD: A webscraping method was used to identify and classify relevant "hits", defined as a state behavioral health webpage with single or multiple references to a study EBPR. A total of 778 hits (unique combinations of webpage and register) were coded. Up to three codes were applied to each hit for the "reasons for the EBPR reference" (EBPR use) dimension, one code was applied to each hit for the "purpose of the EBPR reference" and "intended audience of the webpage containing the hit" dimensions, and up to two codes were applied to each hit for the "funding mentions" dimension. RESULTS: Three EBPRs out of 28 accounted for 73.6% of the hits. The most frequent reason for referencing EBPRs were as a resource for selecting EBPs or validating existing programs and practices. The references tended to appear in reports from the state, in training materials, or guidelines. The references tended to address broad groups of behavioral healthcare professionals. EBPRs were frequently referenced in the context of federal block grants or other federal funding. CONCLUSIONS: Increasing state agencies' awareness and use of the entire range of existing EBPRs may improve implementation of EBPs nationally.


Assuntos
Atenção à Saúde , Prática Clínica Baseada em Evidências , Criança , Humanos , Avaliação de Programas e Projetos de Saúde , Governo Estadual
9.
Bioconjug Chem ; 31(9): 2136-2146, 2020 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-32697078

RESUMO

Engineering cysteines at specific sites in antibodies to create well-defined ADCs for the treatment of cancer is a promising approach to increase the therapeutic index and helps to streamline the manufacturing process. Here, we report the development of an in silico screening procedure to select for optimal sites in an antibody to which a hydrophobic linker-drug can be conjugated. Sites were identified inside the cavity that is naturally present in the Fab part of the antibody. Conjugating a linker-drug to these sites demonstrated the ability of the antibody to shield the hydrophobic character of the linker-drug while resulting ADCs maintained their cytotoxic potency in vitro. Comparison of site-specific ADCs versus randomly conjugated ADCs in an in vivo xenograft model revealed improved efficacy and exposure. We also report a selective reducing agent that is able to reduce the engineered cysteines while leaving the interchain disulfides in the oxidized state. This enables us to manufacture site-specific ADCs without introducing impurities associated with the conventional reduction/oxidation procedure for site-specific conjugation.


Assuntos
Antibióticos Antineoplásicos/química , Cisteína/química , Duocarmicinas/análogos & derivados , Imunoconjugados/química , Animais , Antibióticos Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Duocarmicinas/uso terapêutico , Humanos , Interações Hidrofóbicas e Hidrofílicas , Imunoconjugados/uso terapêutico , Imunoglobulina G/química , Imunoglobulina G/uso terapêutico , Camundongos , Modelos Moleculares , Neoplasias/tratamento farmacológico , Oxirredução
10.
Mol Cancer Ther ; 17(11): 2389-2398, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30093567

RESUMO

Carboxylesterase 1c (CES1c) is responsible for linker-drug instability and poor pharmacokinetics (PK) of several antibody-drug conjugates (ADC) in mice, but not in monkeys or humans. Preclinical development of these ADCs could be improved if the PK in mice would more closely resemble that of humans and is not affected by an enzyme that is irrelevant for humans. SYD985, a HER2-targeting ADC based on trastuzumab and linker-drug vc-seco-DUBA, is also sensitive to CES1c. In the present studies, we first focused on the interaction between CES1c and SYD985 by size- exclusion chromatography, Western blotting, and LC/MS-MS analysis, using recombinant CES1c and plasma samples. Intriguingly, CES1c activity not only results in release of the active toxin DUBA but also in formation of a covalent bond between CES1c and the linker of vc-seco-DUBA. Mass spectrometric studies enabled identification of the CES1c cleavage site on the linker-drug and the structure of the CES1c adduct. To assess the in vivo impact, CES1c-/- SCID mice were generated that showed stable PK for SYD985, comparable to that in monkeys and humans. Patient-derived xenograft (PDX) studies in these mice showed enhanced efficacy compared with PDX studies in CES1c+/+ mice and provided a more accurate prediction of clinical efficacy of SYD985, hence delivering better quality data. It seems reasonable to assume that CES1c-/- SCID mice can increase quality in ADC development much broader for all ADCs that carry linker-drugs susceptible to CES1c, without the need of chemically modifying the linker-drug to specifically increase PK in mice. Mol Cancer Ther; 17(11); 2389-98. ©2018 AACR.


Assuntos
Carboxilesterase/deficiência , Imunoconjugados/farmacologia , Imunoconjugados/farmacocinética , Animais , Carboxilesterase/metabolismo , Domínio Catalítico , Linhagem Celular Tumoral , Feminino , Humanos , Imunoconjugados/química , Camundongos Knockout , Camundongos SCID , Peptídeos/química , Ratos Wistar , Trastuzumab/química , Resultado do Tratamento
11.
Front Physiol ; 9: 207, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29706893

RESUMO

IKr is the rapidly activating component of the delayed rectifier potassium current, the ion current largely responsible for the repolarization of the cardiac action potential. Inherited forms of long QT syndrome (LQTS) (Lees-Miller et al., 1997) in humans are linked to functional modifications in the Kv11.1 (hERG) ion channel and potentially life threatening arrhythmias. There is little doubt now that hERG-related component of IKr in the heart depends on the tetrameric (homo- or hetero-) channels formed by two alternatively processed isoforms of hERG, termed hERG1a and hERG1b. Isoform composition (hERG1a- vs. the b-isoform) has recently been reported to alter pharmacologic responses to some hERG blockers and was proposed to be an essential factor pre-disposing patients for drug-induced QT prolongation. Very little is known about the gating and pharmacological properties of two isoforms in heart membranes. For example, how gating mechanisms of the hERG1a channels differ from that of hERG1b is still unknown. The mechanisms by which hERG 1a/1b hetero-tetramers contribute to function in the heart, or what role hERG1b might play in disease are all questions to be answered. Structurally, the two isoforms differ only in the N-terminal region located in the cytoplasm: hERG1b is 340 residues shorter than hERG1a and the initial 36 residues of hERG1b are unique to this isoform. In this study, we combined electrophysiological measurements for HEK cells, kinetics and structural modeling to tease out the individual contributions of each isoform to Action Potential formation and then make predictions about the effects of having various mixture ratios of the two isoforms. By coupling electrophysiological data with computational kinetic modeling, two proposed mechanisms of hERG gating in two homo-tetramers were examined. Sets of data from various experimental stimulation protocols (HEK cells) were analyzed simultaneously and fitted to Markov-chain models (M-models). The minimization procedure presented here, allowed assessment of suitability of different Markov model topologies and the corresponding parameters that describe the channel kinetics. The kinetics modeling pointed to key differences in the gating kinetics that were linked to the full channel structure. Interactions between soluble domains and the transmembrane part of the channel appeared to be critical determinants of the gating kinetics. The structures of the full channel in the open and closed states were compared for the first time using the recent Cryo-EM resolved structure for full open hERG channel and an homology model for the closed state, based on the highly homolog EAG1 channel. Key potential interactions which emphasize the importance of electrostatic interactions between N-PAS cap, S4-S5, and C-linker are suggested based on the structural analysis. The derived kinetic parameters were later used in higher order models of cells and tissue to track down the effect of varying the ratios of hERG1a and hERG1b on cardiac action potentials and computed electrocardiograms. Simulations suggest that the recovery from inactivation of hERG1b may contribute to its physiologic role of this isoform in the action potential. Finally, the results presented here contribute to the growing body of evidence that hERG1b significantly affects the generation of the cardiac Ikr and plays an important role in cardiac electrophysiology. We highlight the importance of carefully revisiting the Markov models previously proposed in order to properly account for the relative abundance of the hERG1 a- and b- isoforms.

12.
J Comp Neurol ; 525(12): 2735-2781, 2017 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-28510275

RESUMO

The vertebrate retina is a specialized photosensitive tissue comprised of six neuronal and one glial cell types, each of which develops in prescribed proportions at overlapping timepoints from a common progenitor pool. While each of these cells has a specific function contributing to proper vision in the mature animal, their differential representation in the retina as well as the presence of distinctive cellular subtypes makes identifying the transcriptomic signatures that lead to each retinal cell's fate determination and development challenging. We have analyzed transcriptomes from individual cells isolated from the chick retina throughout retinogenesis. While we focused our efforts on the retinal ganglion cells, our transcriptomes of developing chick cells also contained representation from multiple retinal cell types, including photoreceptors and interneurons at different stages of development. Most interesting was the identification of transcriptomes from individual mixed lineage progenitor cells in the chick as these cells offer a window into the cell fate decision-making process. Taken together, these data sets will enable us to uncover the most critical genes acting in the steps of cell fate determination and early differentiation of various retinal cell types.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Neuroglia/metabolismo , Neurônios/metabolismo , Organogênese/fisiologia , Retina/citologia , Retina/embriologia , Células Ganglionares da Retina/metabolismo , Análise de Célula Única/métodos , Células-Tronco/metabolismo , Transcriptoma/fisiologia , Animais , Embrião de Galinha , Perfilação da Expressão Gênica , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/classificação , Células-Tronco/citologia
13.
Mol Cancer Ther ; 15(8): 1900-9, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27256376

RESUMO

Uterine serous carcinoma (USC) is an aggressive form of endometrial cancer. Up to 35% of USC may overexpress the HER2/neu oncogene at strong (i.e., 3+) levels by IHC while an additional 40% to 50% express HER2/neu at moderate (2+) or low (1+) levels. We investigated the efficacy of SYD985, (Synthon Biopharmaceuticals), a novel HER2-targeting antibody-drug conjugate (ADC) composed of the mAb trastuzumab linked to a highly potent DNA-alkylating agent (i.e., duocarmycin) in USC. We also compared the antitumor activity of SYD985 in head-to-head experiments to trastuzumab emtansine (T-DM1), a FDA-approved ADC, against multiple primary USC cell lines expressing different levels of HER2/neu in in vitro and in vivo experiments. Using antibody-dependent cellular cytotoxicity (ADCC), proliferation, viability, and bystander killing assays as well as propidium iodide-based flow cytometry assays and multiple in vivo USC mouse xenograft models, we demonstrate for the first time that SYD985 is a novel ADC with activity against USC with strong (3+) as well as low to moderate (i.e., 1+/2+) HER2/neu expression. SYD985 is 10- to 70-fold more potent than T-DM1 in comparative experiments and, unlike T-DM1, it is active against USC demonstrating moderate/low or heterogeneous HER2/neu expression. Clinical studies with SYD985 in patients harboring chemotherapy-resistant USC with low, moderate, and high HER2 expression are warranted. Mol Cancer Ther; 15(8); 1900-9. ©2016 AACR.


Assuntos
Antineoplásicos/farmacologia , Cistadenocarcinoma Seroso/genética , Expressão Gênica , Imunoconjugados/farmacologia , Indóis , Receptor ErbB-2/antagonistas & inibidores , Neoplasias Uterinas/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Antineoplásicos/química , Efeito Espectador , Catepsina B/genética , Catepsina B/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Classe I de Fosfatidilinositol 3-Quinases , Cistadenocarcinoma Seroso/tratamento farmacológico , Cistadenocarcinoma Seroso/metabolismo , Cistadenocarcinoma Seroso/patologia , Modelos Animais de Doenças , Duocarmicinas , Feminino , Humanos , Imunoconjugados/química , Indóis/química , Camundongos , Pessoa de Meia-Idade , Mutação , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Pirrolidinonas/química , Análise de Sobrevida , Neoplasias Uterinas/tratamento farmacológico , Neoplasias Uterinas/metabolismo , Neoplasias Uterinas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
14.
MAbs ; 8(1): 74-86, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26440530

RESUMO

Monomeric IgA has been proposed as an alternative antibody format for cancer therapy. Here, we present our studies on the production, purification and functional evaluation of anti-HER2 IgA antibodies as anti-cancer agents in comparison to the anti-HER2 IgG1 trastuzumab. MALDI-TOF MS analysis showed profound differences in glycosylation traits across the IgA isotypes and cell lines used for production, including sialylation and linkage thereof, fucosylation (both core and antennary) and the abundance of high-mannose type species. Increases in sialylation proved to positively correlate with in vivo plasma half-lives. The polymerization propensity of anti-HER2 IgA2m2 could be suppressed by an 18-aa deletion of the heavy chain tailpiece - coinciding with the loss of high-mannose type N-glycan species - as well as by 2 cysteine to serine mutations at positions 320 and 480. The HER2 F(ab')2-mediated anti-proliferative effect of the IgA2m1 and IgA2m2 subtypes was similar to IgG1, whereas the IgA1 isotype displayed considerably lower potency and efficacy. The Fc-mediated induction of antibody-dependent cell-mediated cytotoxicity (ADCC) using human whole blood ADCC assays did not demonstrate such clear differences between the IgA isotypes. However, the potency of the anti-HER2 IgA antibodies in these ADCC assays was found to be significantly lower than that of trastuzumab. In vivo anti-tumor activity of the anti-HER2 IgA antibodies was compared to that of trastuzumab in a BT-474 breast cancer xenograft model. Multiple dosing and sialylation of the IgA antibodies compensated for the short in vivo half-life of native IgA antibodies in mice compared to a single dose of IgG1. In the case of the IgA2m2 antibody, the resulting high plasma exposure levels were sufficient to cause clear tumor stasis comparable to that observed for trastuzumab at much lower plasma exposure levels.


Assuntos
Anticorpos Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Ácido N-Acetilneuramínico , Polissacarídeos , Receptor ErbB-2/antagonistas & inibidores , Trastuzumab/farmacologia , Animais , Neoplasias da Mama/sangue , Linhagem Celular Tumoral , Feminino , Glicosilação , Humanos , Imunoglobulina A , Imunoglobulina G , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Mol Cancer Ther ; 14(3): 692-703, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25589493

RESUMO

SYD985 is a HER2-targeting antibody-drug conjugate (ADC) based on trastuzumab and vc-seco-DUBA, a cleavable linker-duocarmycin payload. To evaluate the therapeutic potential of this new ADC, mechanistic in vitro studies and in vivo patient-derived xenograft (PDX) studies were conducted to compare SYD985 head-to-head with T-DM1 (Kadcyla), another trastuzumab-based ADC. SYD985 and T-DM1 had similar binding affinities to HER2 and showed similar internalization. In vitro cytotoxicity assays showed similar potencies and efficacies in HER2 3+ cell lines, but in cell lines with low HER2 expression, SYD985 was 3- to 50-fold more potent than T-DM1. In contrast with T-DM1, SYD985 efficiently induced bystander killing in vitro in HER2-negative (HER2 0) cells mixed with HER2 3+, 2+, or 1+ cell lines. At pH conditions relevant for tumors, cathepsin-B cleavage studies showed efficient release of the active toxin by SYD985 but not by T-DM1. These in vitro data suggest that SYD985 might be a more potent ADC in HER2-expressing tumors in vivo, especially in low HER2-expressing and/or in heterogeneous tumors. In line with this, in vivo antitumor studies in breast cancer PDX models showed that SYD985 is very active in HER2 3+, 2+, and 1+ models, whereas T-DM1 only showed significant antitumor activity in HER2 3+ breast cancer PDX models. These properties of SYD985 may enable expansion of the target population to patients who have low HER2-expressing breast cancer, a patient population with still unmet high medical need.


Assuntos
Anticorpos Monoclonais/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Indóis/farmacologia , Receptor ErbB-2/genética , Animais , Linhagem Celular Tumoral , Duocarmicinas , Feminino , Humanos , Camundongos , Camundongos Nus , Pirrolidinonas/farmacologia , Trastuzumab/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
16.
Mol Pharm ; 12(6): 1813-35, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25635711

RESUMO

Antibody-drug conjugates (ADCs) that are currently on the market or in clinical trials are predominantly based on two drug classes: auristatins and maytansinoids. Both are tubulin binders and block the cell in its progression through mitosis. We set out to develop a new class of linker-drugs based on duocarmycins, potent DNA-alkylating agents that are composed of a DNA-alkylating and a DNA-binding moiety and that bind into the minor groove of DNA. Linker-drugs were evaluated as ADCs by conjugation to the anti-HER2 antibody trastuzumab via reduced interchain disulfides. Duocarmycin 3b, bearing an imidazo[1,2-a]pyridine-based DNA-binding unit, was selected as the drug moiety, notably because of its rapid degradation in plasma. The drug was incorporated into the linker-drugs in its inactive prodrug form, seco-duocarmycin 3a. Linker attachment to the hydroxyl group in the DNA-alkylating moiety was favored over linking to the DNA-binding moiety, as the first approach gave more consistent results for in vitro cytotoxicity and generated ADCs with excellent human plasma stability. Linker-drug 2 was eventually selected based on the properties of the corresponding trastuzumab conjugate, SYD983, which had an average drug-to-antibody ratio (DAR) of about 2. SYD983 showed subnanomolar potencies against multiple human cancer cell lines, was highly efficacious in a BT-474 xenograft model, and had a long half-life in cynomolgus monkeys, in line with high stability in monkey and human plasma. Studies comparing ADCs with a different average DAR showed that a higher average DAR leads to increased efficacy but also to somewhat less favorable physicochemical and toxicological properties. Fractionation of SYD983 with hydrophobic interaction chromatography resulted in SYD985, consisting of about 95% DAR2 and DAR4 species in an approximate 2:1 ratio and having an average DAR of about 2.8. SYD985 combines several favorable properties from the unfractionated ADCs with an improved homogeneity. It was selected for further development and recently entered clinical Phase I evaluation.


Assuntos
Imunoconjugados/química , Indóis/química , Receptor ErbB-2/metabolismo , Antineoplásicos/química , Antineoplásicos/farmacocinética , Linhagem Celular Tumoral , Duocarmicinas , Humanos , Imunoconjugados/farmacocinética , Pirrolidinonas/química
17.
Mol Cancer Ther ; 13(11): 2618-29, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25189543

RESUMO

A linker-drug platform was built on the basis of a cleavable linker-duocarmycin payload for the development of new-generation antibody-drug conjugates (ADC). A leading ADC originating from that platform is SYD983, a HER2-targeting ADC based on trastuzumab. HER2-binding, antibody-dependent cell-mediated cytotoxicity and HER2-mediated internalization are similar for SYD983 as compared with trastuzumab. HER2-expressing cells in vitro are very potently killed by SYD983, but SYD983 is inactive in cells that do not express HER2. SYD983 dose dependently reduces tumor growth in a BT-474 mouse xenograft in vivo. The ADC is stable in human and cynomolgus monkey plasma in vitro but shows relatively poor stability in mouse plasma due to mouse-specific carboxylesterase. SYD983 could be dosed up to 30 mg/kg in cynomolgus monkeys with high exposure, excellent stability in blood, and without severe toxic effects. The monkey safety study showed no SYD983-induced thrombocytopenia and no induction of peripheral sensory neuropathy, both commonly observed in trials and studies with ADCs based on tubulin inhibitors. Finally, to improve homogeneity, SYD983 was further purified by hydrophobic interaction chromatography resulting in an ADC (designated SYD985) predominantly containing DAR2 and DAR4 species. SYD985 showed high antitumor activity in two patient-derived xenograft models of HER2-positive metastatic breast cancers. In conclusion, the data obtained indicate great potential for this new HER2-targeting ADC to become an effective drug for patients with HER2-positive cancers with a favorable safety profile. More generally, this new-generation duocarmycin-based linker-drug technology could be used with other mAbs to serve more indications in oncology.


Assuntos
Imunotoxinas/administração & dosagem , Indóis/administração & dosagem , Receptor ErbB-2/metabolismo , Animais , Células CHO , Linhagem Celular Tumoral , Cricetulus , Duocarmicinas , Feminino , Humanos , Imunotoxinas/química , Indóis/química , Indóis/farmacocinética , Camundongos , Camundongos Endogâmicos BALB C , Terapia de Alvo Molecular , Pirrolidinonas/administração & dosagem , Pirrolidinonas/química , Pirrolidinonas/farmacocinética , Distribuição Aleatória , Ensaios Antitumorais Modelo de Xenoenxerto
18.
J Immunol ; 192(8): 3908-3914, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24639348

RESUMO

Chemokines comprise a family of secreted proteins that activate G protein-coupled chemokine receptors and thereby control the migration of leukocytes during inflammation or immune surveillance. The positional information required for such migratory behavior is governed by the binding of chemokines to membrane-tethered glycosaminoglycans (GAGs), which establishes a chemokine concentration gradient. An often observed but incompletely understood behavior of chemokines is the ability of unrelated chemokines to enhance the potency with which another chemokine subtype can activate its cognate receptor. This phenomenon has been demonstrated to occur between many chemokine combinations and across several model systems and has been dubbed chemokine cooperativity. In this study, we have used GAG binding-deficient chemokine mutants and cell-based functional (migration) assays to demonstrate that chemokine cooperativity is caused by competitive binding of chemokines to GAGs. This mechanistic explanation of chemokine cooperativity provides insight into chemokine gradient formation in the context of inflammation, in which multiple chemokines are secreted simultaneously.


Assuntos
Quimiocinas/metabolismo , Glicosaminoglicanos/metabolismo , Animais , Ligação Competitiva , Células CHO , Quimiocina CCL19/metabolismo , Quimiocina CCL21/metabolismo , Quimiocina CXCL13/metabolismo , Quimiocinas/química , Quimiotaxia , Cricetinae , Cricetulus , Modelos Biológicos , Ligação Proteica , Multimerização Proteica , Receptores de Quimiocinas/metabolismo
19.
J Biol Chem ; 288(10): 7169-81, 2013 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-23341447

RESUMO

Chemokine receptors form a large subfamily of G protein-coupled receptors that predominantly activate heterotrimeric Gi proteins and are involved in immune cell migration. CCX-CKR is an atypical chemokine receptor with high affinity for CCL19, CCL21, and CCL25 chemokines, but is not known to activate intracellular signaling pathways. However, CCX-CKR acts as decoy receptor and efficiently internalizes these chemokines, thereby preventing their interaction with other chemokine receptors, like CCR7 and CCR9. Internalization of fluorescently labeled CCL19 correlated with ß-arrestin2-GFP translocation. Moreover, recruitment of ß-arrestins to CCX-CKR in response to CCL19, CCL21, and CCL25 was demonstrated using enzyme-fragment complementation and bioluminescence resonance energy transfer methods. To unravel why CCX-CKR is unable to activate Gi signaling, CCX-CKR chimeras were constructed by substituting its intracellular loops with the corresponding CCR7 or CCR9 domains. The signaling properties of chimeric CCX-CKR receptors were characterized using a cAMP-responsive element (CRE)-driven reporter gene assay. Unexpectedly, wild type CCX-CKR and a subset of the chimeras induced an increase in CRE activity in response to CCL19, CCL21, and CCL25 in the presence of the Gi inhibitor pertussis toxin. CCX-CKR signaling to CRE required an intact DRY motif. These data suggest that inactive Gi proteins impair CCX-CKR signaling most likely by hindering the interaction of this receptor with pertussis toxin-insensitive G proteins that transduce signaling to CRE. On the other hand, recruitment of the putative signaling scaffold ß-arrestin to CCX-CKR in response to chemokines might allow activation of yet to be identified signal transduction pathways.


Assuntos
Arrestinas/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Receptores CCR/metabolismo , Transdução de Sinais , Animais , Arrestinas/genética , Ligação Competitiva/efeitos dos fármacos , Western Blotting , Células CHO , Linhagem Celular Tumoral , Quimiocina CCL19/metabolismo , Quimiocina CCL19/farmacologia , Quimiocina CCL21/metabolismo , Quimiocina CCL21/farmacologia , Quimiocinas CC/metabolismo , Quimiocinas CC/farmacologia , Cricetinae , Cricetulus , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Microscopia de Fluorescência , Modelos Biológicos , Toxina Pertussis/farmacologia , Ligação Proteica/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Receptores CCR/genética , beta-Arrestinas
20.
Cell Signal ; 25(2): 527-38, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23159578

RESUMO

Parathyroid hormone (PTH) is an anabolic agent that mediates bone formation through activation of the Gα(s)-, Gα(q)- and ß-arrestin-coupled parathyroid hormone receptor type 1 (PTH1R). Pharmacological evidence based on the effect of PTH(7-34), a PTH derivative that is said to preferentially activate ß-arrestin signaling through PTH1R, suggests that PTH1R-activated ß-arrestin signaling mediates anabolic effects on bone. Here, we performed a thorough evaluation of PTH(7-34) signaling behaviour using quantitative assays for ß-arrestin recruitment, Gα(s)- and Gα(q)-signaling. We found that PTH(7-34) inhibited PTH-induced cAMP accumulation, but was unable to induce ß-arrestin recruitment, PTH1R internalization and ERK1/2 phosphorylation in HEK293, CHO and U2OS cells. Thus, the ß-arrestin bias of PTH(7-34) is not apparent in every cell type examined, suggesting that correlating in vivo effects of PTH(7-34) to in vitro pharmacology should be done with caution.


Assuntos
Arrestinas/metabolismo , Hormônio Paratireóideo/farmacologia , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Células CHO , Cálcio/metabolismo , Linhagem Celular , Cricetinae , Cricetulus , AMP Cíclico/metabolismo , Humanos , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Hormônio Paratireóideo/análogos & derivados , Fosforilação , Receptor Tipo 1 de Hormônio Paratireóideo/agonistas , beta-Arrestinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...