Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
ACS Appl Mater Interfaces ; 16(8): 10459-10467, 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38358426

RESUMEN

Inverted colloidal-nanocrystal-based LEDs (NC-LEDs) are highly interesting and invaluable for large-scale display technology and flexible electronics. Semiconductor nanorods (NRs), in addition to the tunable wavelengths of the emitted light (achieved, for example, by the variation of the NR diameter or the diameter of core in a core-shell configuration), also exhibit linearly polarized emission, a larger Stokes shift, faster radiative decay, and slower bleaching kinetics than quantum dots (QDs). Despite these advantages, it is difficult to achieve void-free active NR layers using simple spin-coating techniques. Herein, we employ electrophoretic deposition (EPD) to make closely packed, vertically aligned CdSe/CdS core/shell nanorods (NRs) as the emissive layer. Following an inverted architecture, the device fabricated yields an external quantum efficiency (EQE) of 6.3% and a maximum luminance of 4320 cd/m2 at 11 V. This good performance can be attributed to the vertically aligned NR layer, enhancing the charge transport by reducing the resistance of carrier passage, which is supported by our finite element simulations. To the best of our knowledge, this is the first time vertically aligned NR layers made by EPD have been reported for the fabrication of NC-LEDs and the device performance is one of the best for inverted red NR-LEDs. The findings presented in this work bring forth a simple and effective technique for making vertically aligned NRs, and the mechanism behind the NR-LED device with enhanced performance using these NRs is illustrated. This technique may prove useful to the development of a vast class of nanocrystal-based optoelectronics, including solar cells and laser devices.

2.
Subjectivity ; 15(3): 93-108, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36120081

RESUMEN

The relationship between 'philosophy' and the 'geo' has received renewed attention with the rise of the terrestrial and the planetary as leitmotifs for thinking about the collective subjectivation of particular kinds of world. In some of these conversations, this relationship is developed to consider how social collectives emerge with the production of particular kinds of territorial abstraction. Three decades since Gilles Deleuze and Félix Guattari published What is Philosophy?, book that has a lasting legacy in developing geophilosophy as a particular mode of transcendental empirical enquiry, this special issue revisits the relationship between geophilosophy and the production of an alternative sense of the earth. In this introduction, we approach geophilosophy in its pluralism by showing how the concept does not only concern the question of how to retain a sense of difference and contingency in thought, but also concerns a mode of enquiry that presents opportunities to experiment with alternative forms of collective subjectivation. Assaying the legacy of Deleuze and Guattari's geophilosophy on contemporary forms of earth-thinking, the article identifies the unique demands and geophilosophical possibilities taken up by the contributors to this issue that question how to recuperate another sense of the earth.

3.
Cell Surf ; 7: 100065, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34778603

RESUMEN

The development of new vaccines for TB needs to be underpinned by an understanding of both the molecular and cellular mechanisms of host-pathogen interactions and how the immune response can be modulated to achieve protection from disease. Complement orchestrates many aspects of the innate and adaptive immune responses. However, little is known about the contribution of the complement pathways during TB disease, particularly with respect to mycobacterial phenotype. Extracellular communities (biofilms) of M. tuberculosis are found in the acellular rim of granulomas, during disease, and these are likely to be present in post-primary TB episodes, in necrotic lesions. Our study aimed to determine which mycobacterial cell wall components were altered during biofilm growth and how these cell wall alterations modified the complement response. We have shown that M. tuberculosis biofilms modified their cell wall carbohydrates and elicited reduced classical and lectin pathway activation. Consistent with this finding was the reduction of C3b/iC3b deposition on biofilm cell wall carbohydrate extracts. Here, we have highlighted the role of cell wall carbohydrate alterations during biofilm growth of M. tuberculosis and subsequent modulation of complement activation.

4.
Neoplasia ; 23(2): 210-221, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33385970

RESUMEN

With the recent approval of 3 new antibody drug conjugates (ADCs) for solid tumors, this class of drugs is gaining momentum for the targeted treatment of cancer. Despite significant investment, there are still fundamental issues that are incompletely understood. Three of the recently approved ADCs contain payloads exhibiting bystander effects, where the payload can diffuse out of a targeted cell into adjacent cells. These effects are often studied using a mosaic of antigen positive and negative cells. However, the distance these payloads can diffuse in tumor tissue while maintaining a lethal concentration is unclear. Computational studies suggest bystander effects partially compensate for ADC heterogeneity in tumors in addition to targeting antigen negative cells. However, this type of study is challenging to conduct experimentally due to the low concentrations of extremely potent payloads. In this work, we use a series of 3-dimensional cell culture and primary human tumor xenograft studies to directly track fluorescently labeled ADCs and indirectly follow the payload via an established pharmacodynamic marker (γH2A. X). Using TAK-164, an anti-GCC ADC undergoing clinical evaluation, we show that the lipophilic DNA-alkylating payload, DGN549, penetrates beyond the cell targeted layer in GCC-positive tumor spheroids and primary human tumor xenograft models. The penetration distance is similar to model predictions, where the lipophilicity results in moderate tissue penetration, thereby balancing improved tissue penetration with sufficient cellular uptake to avoid significant washout. These results aid in mechanistic understanding of the interplay between antigen heterogeneity, bystander effects, and heterogeneous delivery of ADCs in the tumor microenvironment to design clinically effective therapeutics.


Asunto(s)
Antineoplásicos Inmunológicos/farmacocinética , Efecto Espectador/efectos de los fármacos , Inmunoconjugados/farmacocinética , Animales , Biomarcadores , Línea Celular Tumoral , Modelos Animales de Enfermedad , Monitoreo de Drogas/métodos , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Fragmentos Fc de Inmunoglobulinas/metabolismo , Ratones , Ratones Transgénicos , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Mol Cancer Ther ; 20(1): 203-212, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33177153

RESUMEN

Several antibody-drug conjugates (ADC) showing strong clinical responses in solid tumors target high expression antigens (HER2, TROP2, Nectin-4, and folate receptor alpha/FRα). Highly expressed tumor antigens often have significant low-level expression in normal tissues, resulting in the potential for target-mediated drug disposition (TMDD) and increased clearance. However, ADCs often do not cross-react with normal tissue in animal models used to test efficacy (typically mice), and the impact of ADC binding to normal tissue antigens on tumor response remains unclear. An antibody that cross-reacts with human and murine FRα was generated and tested in an animal model where the antibody/ADC bind both human tumor FRα and mouse FRα in normal tissue. Previous work has demonstrated that a "carrier" dose of unconjugated antibody can improve the tumor penetration of ADCs with high expression target-antigens. A carrier dose was employed to study the impact on cross-reactive ADC clearance, distribution, and efficacy. Co-administration of unconjugated anti-FRα antibody with the ADC-improved efficacy, even in low expression models where co-administration normally lowers efficacy. By reducing target-antigen-mediated clearance in normal tissue, the co-administered antibody increased systemic exposure, improved tumor tissue penetration, reduced target-antigen-mediated uptake in normal tissue, and increased ADC efficacy. However, payload potency and tumor antigen saturation are also critical to efficacy, as shown with reduced efficacy using too high of a carrier dose. The judicious use of higher antibody doses, either through lower DAR or carrier doses, can improve the therapeutic window by increasing efficacy while lowering target-mediated toxicity in normal tissue.


Asunto(s)
Anticuerpos/administración & dosificación , Anticuerpos/farmacología , Inmunoconjugados/metabolismo , Animales , Anticuerpos/toxicidad , Línea Celular Tumoral , Reacciones Cruzadas/inmunología , Portadores de Fármacos/química , Femenino , Inmunoconjugados/sangre , Ratones , Ratones SCID , Neoplasias/patología , Resultado del Tratamiento
6.
Assist Technol ; 33(4): 178-189, 2021 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-31169462

RESUMEN

Aims: Develop a new pain assessment for youth with communication challenges. The Guard-Putzer Pain Assessment Domains (gPAD) mobile application (app) was designed and tested as a universally accessible way for youth, ages 7-12 years, with a developmental disability (DD) to express their pain experiences through self-report. Methods: A two-phase process developed the design for an app, created an interactive prototype, and tested its face validity and user interface. This work included a comprehensive scoping review of current assessments and pain apps as well as a survey to obtain descriptive data on the clinical practicality of the gPAD to guide the app design. Additionally, 15 therapists reviewed the gPAD assessment. Results: Thirteen respondents (87%) agreed to the statement that they would use the gPAD for this population. School-based practitioners seemed to highlight the most significant needs for the app. Conclusions: Advancement of this app could mainstream the assessment of pain in youth with DD, and other potential populations.


Asunto(s)
Discapacidades del Desarrollo , Aplicaciones Móviles , Adolescente , Niño , Comunicación , Humanos , Dimensión del Dolor , Encuestas y Cuestionarios
7.
Mol Cancer Ther ; 19(10): 2079-2088, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32788205

RESUMEN

Guanylyl cyclase C (GCC) is a unique therapeutic target with expression restricted to the apical side of epithelial cell tight junctions thought to be only accessible by intravenously administered agents on malignant tissues where GCC expression is aberrant. In this study, we sought to evaluate the therapeutic potential of a second-generation investigational antibody-dug conjugate (ADC), TAK-164, comprised of a human anti-GCC mAb conjugated via a peptide linker to the highly cytotoxic DNA alkylator, DGN549. The in vitro binding, payload release, and in vitro activity of TAK-164 was characterized motivating in vivo evaluation. The efficacy of TAK-164 and the relationship to exposure, pharmacodynamic marker activation, and biodistribution was evaluated in xenograft models and primary human tumor xenograft (PHTX) models. We demonstrate TAK-164 selectively binds to, is internalized by, and has potent cytotoxic effects against GCC-expressing cells in vitro A single intravenous administration of TAK-164 (0.76 mg/kg) resulted in significant growth rate inhibition in PHTX models of metastatic colorectal cancer. Furthermore, imaging studies characterized TAK-164 uptake and activity and showed positive relationships between GCC expression and tumor uptake which correlated with antitumor activity. Collectively, our data suggest that TAK-164 is highly active in multiple GCC-positive tumors including those refractory to TAK-264, a GCC-targeted auristatin ADC. A strong relationship between uptake of 89Zr-labeled TAK-164, levels of GCC expression and, most notably, response to TAK-164 therapy in GCC-expressing xenografts and PHTX models. These data supported the clinical development of TAK-164 as part of a first-in-human clinical trial (NCT03449030).


Asunto(s)
Inmunoconjugados/uso terapéutico , Animales , Femenino , Células HEK293 , Humanos , Inmunoconjugados/farmacología , Ratones , Ratones Desnudos , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Cancer Res ; 80(6): 1268-1278, 2020 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-31941698

RESUMEN

Targeted delivery of chemotherapeutics aims to increase efficacy and lower toxicity by concentrating drugs at the site-of-action, a method embodied by the seven current FDA-approved antibody-drug conjugates (ADC). However, a variety of pharmacokinetic challenges result in relatively narrow therapeutic windows for these agents, hampering the development of new drugs. Here, we use a series of prostate-specific membrane antigen-binding single-domain (Humabody) ADC constructs to demonstrate that tissue penetration of protein-drug conjugates plays a major role in therapeutic efficacy. Counterintuitively, a construct with lower in vitro potency resulted in higher in vivo efficacy than other protein-drug conjugates. Biodistribution data, tumor histology images, spheroid experiments, in vivo single-cell measurements, and computational results demonstrate that a smaller size and slower internalization rate enabled higher tissue penetration and more cell killing. The results also illustrate the benefits of linking an albumin-binding domain to the single-domain ADCs. A construct lacking an albumin-binding domain was rapidly cleared, leading to lower tumor uptake (%ID/g) and decreased in vivo efficacy. In conclusion, these results provide evidence that reaching the maximum number of cells with a lethal payload dose correlates more strongly with in vivo efficacy than total tumor uptake or in vitro potency alone for these protein-drug conjugates. Computational modeling and protein engineering can be used to custom design an optimal framework for controlling internalization, clearance, and tissue penetration to maximize cell killing. SIGNIFICANCE: A mechanistic study of protein-drug conjugates demonstrates that a lower potency compound is more effective in vivo than other agents with equal tumor uptake due to improved tissue penetration and cellular distribution.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Inmunoconjugados/farmacocinética , Modelos Biológicos , Neoplasias de la Próstata/tratamiento farmacológico , Anticuerpos de Dominio Único/farmacología , Animales , Antineoplásicos Alquilantes/química , Antineoplásicos Alquilantes/uso terapéutico , Línea Celular Tumoral , Simulación por Computador , Femenino , Humanos , Inmunoconjugados/química , Inmunoconjugados/uso terapéutico , Masculino , Ratones , Microscopía Confocal , Neoplasias de la Próstata/diagnóstico por imagen , Neoplasias de la Próstata/patología , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/uso terapéutico , Esferoides Celulares , Relación Estructura-Actividad , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Bioconjug Chem ; 31(1): 93-103, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31747250

RESUMEN

Antibody-drug conjugates have elicited great interest recently as targeted chemotherapies for cancer. Recent preclinical and clinical data have continued to raise questions about optimizing the design of these complex therapeutics. Biochemical methods for site-specific antibody conjugation have been a design feature of recent clinical ADCs, and preclinical reports suggest that site-specifically conjugated ADCs generically offer improved therapeutic indices (i.e., the fold difference between efficacious and maximum tolerated doses). Here we present the results of a systematic preclinical comparison of ADCs embodying the DNA-alkylating linker-payload DGN549 generated with both heterogeneous lysine-directed and site-specific cysteine-directed conjugation chemistries. Importantly, the catabolites generated by each ADC are the same regardless of the conjugation format. In two different model systems evaluated, the site-specific ADC showed a therapeutic index benefit. However, the therapeutic index benefit is different in each case: both show evidence of improved tolerability, though with different magnitudes, and in one case significant efficacy improvement is also observed. These results support our contention that conjugation chemistry of ADCs is best evaluated in the context of a particular antibody, target, and linker-payload, and ideally across multiple disease models.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Benzodiazepinas/uso terapéutico , Inmunoconjugados/uso terapéutico , Lisina/uso terapéutico , Neoplasias/tratamiento farmacológico , Oxindoles/uso terapéutico , Animales , Antineoplásicos Alquilantes/efectos adversos , Antineoplásicos Alquilantes/química , Antineoplásicos Alquilantes/farmacocinética , Antineoplásicos Alquilantes/uso terapéutico , Antineoplásicos Inmunológicos/efectos adversos , Antineoplásicos Inmunológicos/química , Antineoplásicos Inmunológicos/farmacocinética , Benzodiazepinas/efectos adversos , Benzodiazepinas/química , Benzodiazepinas/farmacocinética , Línea Celular Tumoral , Femenino , Humanos , Inmunoconjugados/efectos adversos , Inmunoconjugados/química , Inmunoconjugados/farmacocinética , Lisina/efectos adversos , Lisina/química , Lisina/farmacocinética , Ratones , Ratones SCID , Oxindoles/efectos adversos , Oxindoles/química , Oxindoles/farmacocinética , Índice Terapéutico
10.
Mol Pharm ; 17(1): 50-58, 2020 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-31742408

RESUMEN

DNA-targeting indolinobenzodiazepine dimer (IGN) payloads are used in several clinical-stage antibody-drug conjugates. IGN drugs alkylate DNA through the single imine moiety present in the dimer in contrast to the pyrrolobenzodiazepine dimer drugs, such as talirine and tesirine, which contain two imine moieties per dimer and cross-link DNA. This study explored the mechanism of binding of IGN to DNA in cells and to synthetic duplex and hairpin oligonucleotides. New, highly sensitive IGN-DNA binding enzyme-linked immunosorbent assay methods were developed using biotinylated IGN analogues (monoimine, diimine, and diamine IGNs) and digoxigenin-labeled duplex oligonucleotides, which allowed the measurement of drug-DNA adducts in viable cells at concentrations below IC50. Furthermore, the release of free drug from the IGN-DNA adduct upon treatment with nuclease ex vivo was tested under physiological conditions. The monoimine IGN drug formed a highly stable adduct with DNA in cells, with stability similar to that of the diimine drug analogue. Both monoimine and diimine IGN-DNA adducts released free drugs upon DNA cleavage by nuclease at 37 °C, although more free drug was released from the monoimine compared to the diimine adduct, which presumably was partly cross-linked. The strong binding of the monoimine IGN drug to duplex DNA results from both the noncovalent IGN-DNA interaction and the covalent bond formation between the 2-amino group of a guanine residue and the imine moiety in IGN.


Asunto(s)
Antineoplásicos/química , Benzodiazepinas/química , Aductos de ADN/química , ADN/química , Inmunoconjugados/farmacología , Indoles/química , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Línea Celular Tumoral , Aductos de ADN/metabolismo , Dimerización , Ensayo de Immunospot Ligado a Enzimas , Humanos , Iminas/química , Inmunoconjugados/administración & dosificación , Oligonucleótidos/química , Pirroles/química
11.
ACS Med Chem Lett ; 10(10): 1386-1392, 2019 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-31620223

RESUMEN

Antibody-drug conjugates (ADCs) that incorporate the exatecan derivative DXd in their payload are showing promising clinical results in solid tumor indications. The payload has an F-ring that also contains a second chiral center, both of which complicate its synthesis and derivatization. Here we report on new camptothecin-ADCs that do not have an F-ring in their payloads yet behave similarly to DXd-bearing conjugates in vitro and in vivo. This simplification allows easier derivatization of camptothecin A and B rings for structure-activity relationship studies and payload optimization. ADCs having different degrees of bystander killing and the ability to release hydroxyl or thiol-bearing metabolites following peptide linker cleavage were investigated.

12.
Mol Pharm ; 16(12): 4817-4825, 2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31609629

RESUMEN

Although peptide linkers are used in multiple clinical-stage ADCs, there are only few reports on optimizing peptide linkers for efficient lysosomal proteolysis and for stability in circulation. We screened multiple dipeptide linkers for efficiency of proteolysis and compared them to the dipeptide linkers currently being evaluated in the clinic: Val-Cit, Val-Ala, and Ala-Ala. Lead dipeptide linkers selected from the initial screen were incorporated into ADCs with indolinobenzodiazepine dimer (IGN) payloads to evaluate cellular processing, in vitro cytotoxic activity, plasma stability, and in vivo efficacy. ADCs with several dipeptide linkers bearing l-amino acids showed faster lysosomal processing in target cancer cells compared to the l-Ala-l-Ala linked ADC. These variances in linker processing rates did not result in different in vitro and in vivo activities among peptide linker ADCs, presumably due to accumulation of threshold cytotoxic catabolite levels for ADCs of several peptide linkers in the cell lines and xenografts tested. ADCs with l-amino acid dipeptide linkers exhibited superior in vitro cytotoxic potencies in multiple cell lines compared to an ADC with a d-Ala-d-Ala dipeptide linker and an ADC with a noncleavable linker. This work adds to the toolbox of stable, lysosomally cleavable peptide linkers for ADCs.


Asunto(s)
Anticuerpos/química , Biopolímeros/química , Dipéptidos/química , Inmunoconjugados/química , Lisosomas/metabolismo , Animales , Antineoplásicos/química , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Humanos , Ratones , Ratones SCID , Estructura Molecular , Ensayos Antitumor por Modelo de Xenoinjerto
13.
ACS Med Chem Lett ; 10(8): 1211-1215, 2019 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-31413807

RESUMEN

Indolinobenzodiazepine DNA alkylators (IGNs) are the cytotoxic payloads in antibody-drug conjugates (ADCs) currently undergoing Phase I clinical evaluation (IMGN779, IMGN632, and TAK164). These ADCs possess linkers that have been incorporated into a central substituted phenyl spacer. Here, we present an alternative strategy for the IGNs, linking through a carbamate at the readily available N-10 amine present in the monoimine containing dimer. As a result, we have designed a series of N-10 linked IGN ADCs with a wide range of in vitro potency and tolerability, which may allow us to better match an IGN with a particular target based on the potential dosing needs.

14.
Mol Pharm ; 16(9): 3926-3937, 2019 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-31287952

RESUMEN

Antibody-drug conjugates are an emerging class of cancer therapeutics constructed from monoclonal antibodies conjugated with small molecule effectors. First-generation molecules of this class often employed heterogeneous conjugation chemistry, but many site-specifically conjugated ADCs have been described recently. Here, we undertake a systematic comparison of ADCs made with the same antibody and the same macrocyclic maytansinoid effector but conjugated either heterogeneously at lysine residues or site-specifically at cysteine residues. Characterization of these ADCs in vitro reveals generally similar properties, including a similar catabolite profile, a key element in making a meaningful comparison of conjugation chemistries. In a mouse model of cervical cancer, the lysine-conjugated ADC affords greater efficacy on a molar payload basis. Rather than making general conclusions about ADCs conjugated by a particular chemistry, we interpret these results as highlighting the complexity of ADCs and the interplay between payload class, linker chemistry, target antigen, and other variables that determine efficacy in a given setting.


Asunto(s)
Anticuerpos Monoclonales/química , Cisteína/química , Inmunoconjugados/farmacocinética , Inmunoconjugados/uso terapéutico , Lisina/química , Maitansina/inmunología , Neoplasias del Cuello Uterino/tratamiento farmacológico , Animales , Supervivencia Celular/efectos de los fármacos , Femenino , Células HeLa , Humanos , Inmunoconjugados/administración & dosificación , Inyecciones Intravenosas , Ratones , Ratones SCID , Resultado del Tratamiento , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Mol Cancer Ther ; 17(3): 650-660, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29440292

RESUMEN

Tumor-selective delivery of cytotoxic agents in the form of antibody-drug conjugates (ADCs) is now a clinically validated approach for cancer treatment. In an attempt to improve the clinical success rate of ADCs, emphasis has been recently placed on the use of DNA-cross-linking pyrrolobenzodiazepine compounds as the payload. Despite promising early clinical results with this class of ADCs, doses achievable have been low due to systemic toxicity. Here, we describe the development of a new class of potent DNA-interacting agents wherein changing the mechanism of action from a cross-linker to a DNA alkylator improves the tolerability of the ADC. ADCs containing the DNA alkylator displayed similar in vitro potency, but improved bystander killing and in vivo efficacy, compared with those of the cross-linker. Thus, the improved in vivo tolerability and antitumor activity achieved in rodent models with ADCs of the novel DNA alkylator could provide an efficacious, yet safer option for cancer treatment. Mol Cancer Ther; 17(3); 650-60. ©2018 AACR.


Asunto(s)
Inmunoconjugados/farmacología , Sustancias Intercalantes/farmacología , Neoplasias/tratamiento farmacológico , Índice Terapéutico de los Medicamentos , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Antineoplásicos Alquilantes/química , Antineoplásicos Alquilantes/metabolismo , Antineoplásicos Alquilantes/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Reactivos de Enlaces Cruzados/química , ADN/genética , ADN/metabolismo , Diseño de Fármacos , Humanos , Inmunoconjugados/química , Inmunoconjugados/metabolismo , Sustancias Intercalantes/química , Sustancias Intercalantes/metabolismo , Ratones , Neoplasias/patología , Carga Tumoral/efectos de los fármacos
16.
J Oncol Pract ; 13(10): 693-699, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28915075

RESUMEN

Physician aid in dying (PAD) or assisted suicide is becoming legal in more US jurisdictions. Meanwhile, the needs of terminally ill patients with cancer are receiving greater attention, including the integration of palliative care into oncology practice. This article highlights a case vignette of a patient with advanced cancer who requests PAD from her oncologist, as a backdrop to help the practicing oncologist examine his or her moral stance regarding participation in aid in dying. The article concludes by offering a framework within which the practicing oncologist can receive and process a patient's request for PAD.


Asunto(s)
Neoplasias/terapia , Oncólogos , Relaciones Médico-Paciente , Suicidio Asistido , Actitud del Personal de Salud , Beneficencia , Humanos , Cuidados Paliativos , Prioridad del Paciente , Autonomía Personal , Rol del Médico , Suicidio Asistido/ética , Suicidio Asistido/legislación & jurisprudencia , Enfermo Terminal , Estados Unidos
17.
Bioconjug Chem ; 28(5): 1371-1381, 2017 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-28388844

RESUMEN

Antibody-drug conjugates (ADCs) are being actively pursued as a treatment option for cancer following the regulatory approval of brentuximab vedotin (Adcetris) and ado-trastuzumab emtansine (Kadcyla). ADCs consist of a cytotoxic agent conjugated to a targeting antibody through a linker. The two approved ADCs (and most ADCs now in the clinic that use a microtubule disrupting agent as the payload) are heterogeneous conjugates with an average drug-to-antibody ratio (DAR) of 3-4 (potentially ranging from 0 to 8 for individual species). Ado-trastuzumab emtansine employs DM1, a semisynthetic cytotoxic payload of the maytansinoid class, which is conjugated via lysine residues of the antibody to an average DAR of 3.5. To understand the effect of DAR on the preclinical properties of ADCs using maytansinoid cytotoxic agents, we prepared a series of conjugates with a cleavable linker (M9346A-sulfo-SPDB-DM4 targeting folate receptor α (FRα)) or an uncleavable linker (J2898A-SMCC-DM1 targeting the epidermal growth factor receptor (EGFR)) with varying DAR and evaluated their biochemical characteristics, in vivo stability, efficacy, and tolerability. For both formats, a series of ADCs with DARs ranging from low (average of ∼2 and range of 0-4) to very high (average of 10 and range of 7-14) were prepared in good yield with high monomer content and low levels of free cytotoxic agent. The in vitro potency consistently increased with increasing DAR at a constant antibody concentration. We then characterized the in vivo disposition of these ADCs. Pharmacokinetic analysis showed that conjugates with an average DAR below ∼6 had comparable clearance rates, but for those with an average DAR of ∼9-10, rapid clearance was observed. Biodistribution studies in mice showed that these 9-10 DAR ADCs rapidly accumulate in the liver, with maximum localization for this organ at 24-28% percentage injected dose per gram (%ID/g) compared with 7-10% for lower-DAR conjugates (all at 2-6 h post-injection). Our preclinical findings on tolerability and efficacy suggest that maytansinoid conjugates with DAR ranging from 2 to 6 have a better therapeutic index than conjugates with very high DAR (∼9-10). These very high DAR ADCs suffer from decreased efficacy, likely due to faster clearance. These results support the use of DAR 3-4 for maytansinoid ADCs but suggest that the exploration of lower or higher DAR may be warranted depending on the biology of the target antigen.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Antineoplásicos Fitogénicos/farmacocinética , Inmunoconjugados/farmacocinética , Maitansina/farmacocinética , Animales , Antineoplásicos Fitogénicos/farmacología , Femenino , Humanos , Inmunoconjugados/farmacología , Células KB , Maitansina/farmacología , Ratones , Distribución Tisular , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Bioconjug Chem ; 27(7): 1588-98, 2016 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-27174129

RESUMEN

Antibody-drug conjugates (ADCs) have become a widely investigated modality for cancer therapy, in part due to the clinical findings with ado-trastuzumab emtansine (Kadcyla). Ado-trastuzumab emtansine utilizes the Ab-SMCC-DM1 format, in which the thiol-functionalized maytansinoid cytotoxic agent, DM1, is linked to the antibody (Ab) via the maleimide moiety of the heterobifunctional SMCC linker. The pharmacokinetic (PK) data for ado-trastuzumab emtansine point to a faster clearance for the ADC than for total antibody. Cytotoxic agent release in plasma has been reported with nonmaytansinoid, cysteine-linked ADCs via thiol-maleimide exchange, for example, brentuximab vedotin. For Ab-SMCC-DM1 ADCs, however, the main catabolite reported is lysine-SMCC-DM1, the expected product of intracellular antibody proteolysis. To understand these observations better, we conducted a series of studies to examine the stability of the thiol-maleimide linkage, utilizing the EGFR-targeting conjugate, J2898A-SMCC-DM1, and comparing it with a control ADC made with a noncleavable linker that lacked a thiol-maleimide adduct (J2898A-(CH2)3-DM). We employed radiolabeled ADCs to directly measure both the antibody and the ADC components in plasma. The PK properties of the conjugated antibody moiety of the two conjugates, J2898A-SMCC-DM1 and J2898A-(CH2)3-DM (each with an average of 3.0 to 3.4 maytansinoid molecules per antibody), appear to be similar to that of the unconjugated antibody. Clearance values of the intact conjugates were slightly faster than those of the Ab components. Furthermore, J2898A-SMCC-DM1 clears slightly faster than J2898A-(CH2)3-DM, suggesting that there is a fraction of maytansinoid loss from the SMCC-DM1 ADC, possibly through a thiol-maleimide dependent mechanism. Experiments on ex vivo stability confirm that some loss of maytansinoid from Ab-SMCC-DM1 conjugates can occur via thiol elimination, but at a slower rate than the corresponding rate of loss reported for thiol-maleimide links formed at thiols derived by reduction of endogenous cysteine residues in antibodies, consistent with expected differences in thiol-maleimide stability related to thiol pKa. These findings inform the design strategy for future ADCs.


Asunto(s)
Inmunoconjugados/química , Inmunoconjugados/farmacocinética , Lisina/química , Maleimidas/química , Maitansina/química , Animales , Estabilidad de Medicamentos , Ratones , Relación Estructura-Actividad
19.
Antimicrob Agents Chemother ; 60(3): 1349-59, 2015 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-26666933

RESUMEN

This study examined the activity of the novel antimicrobial combination ceftazidime-avibactam against Enterobacteriaceae exhibiting different outer membrane permeability profiles, specifically with or without porins and with or without expression of the main efflux pump (AcrAB-TolC). The addition of the outer membrane permeabilizer polymyxin B nonapeptide increased the antibacterial activities of avibactam alone, ceftazidime alone, and ceftazidime-avibactam against the characterized clinical isolates of Escherichia coli, Enterobacter aerogenes, and Klebsiella pneumoniae. This enhancement of activities was mainly due to increased passive penetration of compounds since inhibition of efflux by the addition of phenylalanine-arginine ß-naphthylamide affected the MICs minimally. OmpF (OmpK35) or OmpC (OmpK36) pores were not the major route by which avibactam crossed the outer membranes of E. coli and K. pneumoniae. In contrast, Omp35 and Omp36 allowed diffusion of avibactam across the outer membrane of E. aerogenes, although other diffusion channels for avibactam were also present in that species. It was clear that outer membrane permeability and outer membrane pore-forming proteins play a key role in the activity of ceftazidime-avibactam. Nevertheless, the MICs of ceftazidime-avibactam (with 4 mg/liter avibactam) against the ceftazidime-resistant clinical isolates of the three species of Enterobacteriaceae studied were ≤ 8 mg/liter, regardless of outer membrane permeability changes resulting from an absence of defined porin proteins or upregulation of efflux.


Asunto(s)
Antibacterianos/farmacología , Compuestos de Azabiciclo/farmacología , Proteínas de la Membrana Bacteriana Externa/metabolismo , Ceftazidima/farmacología , Enterobacteriaceae/efectos de los fármacos , Porinas/metabolismo , Proteínas de la Membrana Bacteriana Externa/genética , Permeabilidad de la Membrana Celular/efectos de los fármacos , Combinación de Medicamentos , Enterobacter aerogenes/efectos de los fármacos , Enterobacteriaceae/genética , Enterobacteriaceae/metabolismo , Escherichia coli/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Fenilalanina/análogos & derivados , Fenilalanina/farmacología , Polimixina B/farmacología , Porinas/genética
20.
Antimicrob Agents Chemother ; 59(6): 3623-6, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25824208

RESUMEN

The mechanism of aminoglycoside resistance among 338 gentamicin-nonsusceptible Gram-negative bacteria (207 Enterobacteriaceae and 131 Pseudomonas aeruginosa) was assessed, and the in vitro activity of ceftazidime-avibactam against these isolates was determined. Aminoglycoside-modifying enzymes were detected in 91.8% of Enterobacteriaceae and 13.7% of P. aeruginosa isolates. A single strain of Klebsiella pneumoniae harbored a 16S rRNA methylase (ArmA). The ceftazidime-avibactam MIC90 values were 0.5 µg/ml (MIC, ≤8 µg/ml for 100% of isolates) and 16 µg/ml (MIC, ≤8 µg/ml for 87.8% of isolates) against gentamicin-nonsusceptible Enterobacteriaceae and P. aeruginosa isolates, respectively.


Asunto(s)
Antibacterianos/farmacología , Compuestos de Azabiciclo/farmacología , Ceftazidima/farmacología , Gentamicinas/farmacología , Bacterias Gramnegativas/efectos de los fármacos , Canadá , Combinación de Medicamentos , Hospitales , Humanos , Pruebas de Sensibilidad Microbiana , ARN Ribosómico 16S/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA