Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Annu Rev Immunol ; 36: 695-715, 2018 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-29490163

RESUMEN

The unique class of heavy chain-only antibodies, present in Camelidae, can be shrunk to just the variable region of the heavy chain to yield VHHs, also called nanobodies. About one-tenth the size of their full-size counterparts, nanobodies can serve in applications similar to those for conventional antibodies, but they come with a number of signature advantages that find increasing application in biology. They not only function as crystallization chaperones but also can be expressed inside cells as such, or fused to other proteins to perturb the function of their targets, for example, by enforcing their localization or degradation. Their small size also affords advantages when applied in vivo, for example, in imaging applications. Here we review such applications, with particular emphasis on those areas where conventional antibodies would face a more challenging environment.


Asunto(s)
Anticuerpos de Dominio Único/genética , Anticuerpos de Dominio Único/inmunología , Animales , Formación de Anticuerpos , Técnicas de Visualización de Superficie Celular , Ingeniería Genética , Humanos , Cadenas Pesadas de Inmunoglobulina/biosíntesis , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Pesadas de Inmunoglobulina/inmunología , Anticuerpos de Dominio Único/biosíntesis , Anticuerpos de Dominio Único/uso terapéutico , Relación Estructura-Actividad
2.
Annu Rev Cell Dev Biol ; 34: 163-188, 2018 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-30110557

RESUMEN

Molecular biologists and chemists alike have long sought to modify proteins with substituents that cannot be installed by standard or even advanced genetic approaches. We here describe the use of transpeptidases to achieve these goals. Living systems encode a variety of transpeptidases and peptide ligases that allow for the enzyme-catalyzed formation of peptide bonds, and protein engineers have used directed evolution to enhance these enzymes for biological applications. We focus primarily on the transpeptidase sortase A, which has become popular over the past few years for its ability to perform a remarkably wide variety of protein modifications, both in vitro and in living cells.


Asunto(s)
Aminoaciltransferasas/genética , Proteínas Bacterianas/genética , Cisteína Endopeptidasas/genética , Péptidos/genética , Peptidil Transferasas/genética , Secuencia de Aminoácidos/genética , Aminoaciltransferasas/química , Proteínas Bacterianas/química , Catálisis , Cisteína Endopeptidasas/química , Humanos , Péptidos/química , Peptidil Transferasas/química , Ingeniería de Proteínas , Especificidad por Sustrato
3.
Proc Natl Acad Sci U S A ; 116(28): 14181-14190, 2019 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-31068469

RESUMEN

Extracellular matrix (ECM) deposition is a hallmark of many diseases, including cancer and fibroses. To exploit the ECM as an imaging and therapeutic target, we developed alpaca-derived libraries of "nanobodies" against disease-associated ECM proteins. We describe here one such nanobody, NJB2, specific for an alternatively spliced domain of fibronectin expressed in disease ECM and neovasculature. We showed by noninvasive in vivo immuno-PET/CT imaging that NJB2 detects primary tumors and metastatic sites with excellent specificity in multiple models of breast cancer, including human and mouse triple-negative breast cancer, and in melanoma. We also imaged mice with pancreatic ductal adenocarcinoma (PDAC) in which NJB2 was able to detect not only PDAC tumors but also early pancreatic lesions called pancreatic intraepithelial neoplasias, which are challenging to detect by any current imaging modalities, with excellent clarity and signal-to-noise ratios that outperformed conventional 2-fluorodeoxyglucose PET/CT imaging. NJB2 also detected pulmonary fibrosis in a bleomycin-induced fibrosis model. We propose NJB2 and similar anti-ECM nanobodies as powerful tools for noninvasive detection of tumors, metastatic lesions, and fibroses. Furthermore, the selective recognition of disease tissues makes NJB2 a promising candidate for nanobody-based therapeutic applications.


Asunto(s)
Carcinogénesis/genética , Carcinoma Ductal Pancreático/diagnóstico por imagen , Matriz Extracelular/efectos de los fármacos , Neoplasias Pancreáticas/diagnóstico por imagen , Animales , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/patología , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Femenino , Fibrosis/patología , Humanos , Masculino , Ratones , Neoplasias Pancreáticas/patología , Tomografía Computarizada por Tomografía de Emisión de Positrones , Neoplasias de la Próstata/diagnóstico por imagen , Neoplasias de la Próstata/patología , Radiofármacos/farmacología , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/farmacología , Neoplasias Pancreáticas
4.
Proc Natl Acad Sci U S A ; 115(15): 3912-3917, 2018 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-29581255

RESUMEN

Ipilimumab, a monoclonal antibody that recognizes cytotoxic T lymphocyte antigen (CTLA)-4, was the first approved "checkpoint"-blocking anticancer therapy. In mouse tumor models, the response to antibodies against CTLA-4 depends entirely on expression of the Fcγ receptor (FcγR), which may facilitate antibody-dependent cellular phagocytosis, but the contribution of simple CTLA-4 blockade remains unknown. To understand the role of CTLA-4 blockade in the complete absence of Fc-dependent functions, we developed H11, a high-affinity alpaca heavy chain-only antibody fragment (VHH) against CTLA-4. The VHH H11 lacks an Fc portion, binds monovalently to CTLA-4, and inhibits interactions between CTLA-4 and its ligand by occluding the ligand-binding motif on CTLA-4 as shown crystallographically. We used H11 to visualize CTLA-4 expression in vivo using whole-animal immuno-PET, finding that surface-accessible CTLA-4 is largely confined to the tumor microenvironment. Despite this, H11-mediated CTLA-4 blockade has minimal effects on antitumor responses. Installation of the murine IgG2a constant region on H11 dramatically enhances its antitumor response. Coadministration of the monovalent H11 VHH blocks the efficacy of a full-sized therapeutic antibody. We were thus able to demonstrate that CTLA-4-binding antibodies require an Fc domain for antitumor effect.


Asunto(s)
Antígeno CTLA-4/inmunología , Fragmentos Fc de Inmunoglobulinas/administración & dosificación , Fragmentos de Inmunoglobulinas/administración & dosificación , Neoplasias/terapia , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Antígeno CTLA-4/química , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Fragmentos Fc de Inmunoglobulinas/química , Fragmentos Fc de Inmunoglobulinas/inmunología , Fragmentos de Inmunoglobulinas/química , Fragmentos de Inmunoglobulinas/inmunología , Inmunoglobulina G/administración & dosificación , Inmunoglobulina G/inmunología , Inmunoterapia , Ratones , Ratones Endogámicos C57BL , Neoplasias/inmunología , Dominios Proteicos
5.
Proc Natl Acad Sci U S A ; 114(38): 10184-10189, 2017 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-28874561

RESUMEN

CD47 is an antiphagocytic ligand broadly expressed on normal and malignant tissues that delivers an inhibitory signal through the receptor signal regulatory protein alpha (SIRPα). Inhibitors of the CD47-SIRPα interaction improve antitumor antibody responses by enhancing antibody-dependent cellular phagocytosis (ADCP) in xenograft models. Endogenous expression of CD47 on a variety of cell types, including erythrocytes, creates a formidable antigen sink that may limit the efficacy of CD47-targeting therapies. We generated a nanobody, A4, that blocks the CD47-SIRPα interaction. A4 synergizes with anti-PD-L1, but not anti-CTLA4, therapy in the syngeneic B16F10 melanoma model. Neither increased dosing nor half-life extension by fusion of A4 to IgG2a Fc (A4Fc) overcame the issue of an antigen sink or, in the case of A4Fc, systemic toxicity. Generation of a B16F10 cell line that secretes the A4 nanobody showed that an enhanced response to several immune therapies requires near-complete blockade of CD47 in the tumor microenvironment. Thus, strategies to localize CD47 blockade to tumors may be particularly valuable for immune therapy.


Asunto(s)
Antígeno CD47/antagonistas & inhibidores , Inmunoterapia/métodos , Melanoma Experimental/terapia , Anticuerpos de Dominio Único/uso terapéutico , Anemia/inducido químicamente , Animales , Antígeno CD47/inmunología , Evaluación Preclínica de Medicamentos , Ratones Endogámicos C57BL , Fagocitosis , Anticuerpos de Dominio Único/inmunología , Anticuerpos de Dominio Único/farmacología , Microambiente Tumoral
6.
Nature ; 503(7476): 406-9, 2013 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-24141948

RESUMEN

Influenza A virus-specific B lymphocytes and the antibodies they produce protect against infection. However, the outcome of interactions between an influenza haemagglutinin-specific B cell via its receptor (BCR) and virus is unclear. Through somatic cell nuclear transfer we generated mice that harbour B cells with a BCR specific for the haemagglutinin of influenza A/WSN/33 virus (FluBI mice). Their B cells secrete an immunoglobulin gamma 2b that neutralizes infectious virus. Whereas B cells from FluBI and control mice bind equivalent amounts of virus through interaction of haemagglutinin with surface-disposed sialic acids, the A/WSN/33 virus infects only the haemagglutinin-specific B cells. Mere binding of virus is not sufficient for infection of B cells: this requires interactions of the BCR with haemagglutinin, causing both disruption of antibody secretion and FluBI B-cell death within 18 h. In mice infected with A/WSN/33, lung-resident FluBI B cells are infected by the virus, thus delaying the onset of protective antibody release into the lungs, whereas FluBI cells in the draining lymph node are not infected and proliferate. We propose that influenza targets and kills influenza-specific B cells in the lung, thus allowing the virus to gain purchase before the initiation of an effective adaptive response.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/virología , Orthomyxoviridae/fisiología , Receptores de Antígenos de Linfocitos B/inmunología , Animales , Anticuerpos/inmunología , Anticuerpos/metabolismo , Especificidad de Anticuerpos/inmunología , Linfocitos B/metabolismo , Linfocitos B/patología , Muerte Celular , Femenino , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Pulmón/citología , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/virología , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Pruebas de Neutralización , Técnicas de Transferencia Nuclear , Orthomyxoviridae/patogenicidad , Receptores de Antígenos de Linfocitos B/metabolismo , Replicación Viral
7.
Proc Natl Acad Sci U S A ; 113(19): E2646-54, 2016 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-27091975

RESUMEN

Therapeutic antitumor antibodies treat cancer by mobilizing both innate and adaptive immunity. CD47 is an antiphagocytic ligand exploited by tumor cells to blunt antibody effector functions by transmitting an inhibitory signal through its receptor signal regulatory protein alpha (SIRPα). Interference with the CD47-SIRPα interaction synergizes with tumor-specific monoclonal antibodies to eliminate human tumor xenografts by enhancing macrophage-mediated antibody-dependent cellular phagocytosis (ADCP), but synergy between CD47 blockade and ADCP has yet to be demonstrated in immunocompetent hosts. Here, we show that CD47 blockade alone or in combination with a tumor-specific antibody fails to generate antitumor immunity against syngeneic B16F10 tumors in mice. Durable tumor immunity required programmed death-ligand 1 (PD-L1) blockade in combination with an antitumor antibody, with incorporation of CD47 antagonism substantially improving response rates. Our results highlight an underappreciated contribution of the adaptive immune system to anti-CD47 adjuvant therapy and suggest that targeting both innate and adaptive immune checkpoints can potentiate the vaccinal effect of antitumor antibody therapy.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/administración & dosificación , Antígeno CD47/efectos de los fármacos , Antígeno CD47/inmunología , Vacunas contra el Cáncer/administración & dosificación , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/terapia , Inmunidad Adaptativa/efectos de los fármacos , Inmunidad Adaptativa/inmunología , Animales , Anticuerpos Monoclonales de Origen Murino/inmunología , Antineoplásicos/administración & dosificación , Antineoplásicos/inmunología , Vacunas contra el Cáncer/inmunología , Femenino , Inmunización/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Terapia Molecular Dirigida/métodos , Neoplasias Experimentales/patología , Resultado del Tratamiento
8.
J Immunol ; 197(12): 4838-4847, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27821668

RESUMEN

mAbs specific for surface proteins on APCs can serve as Ag-delivery vehicles that enhance immunogenicity. The practical use of such constructs is limited by the challenge of expressing and modifying full-sized mAbs. We generated single-domain Ab fragments (VHHs) specific for class II MHC (MHCII), CD11b, and CD36. VHH sequences were modified by inclusion of a C-terminal sortase motif to allow site-specific conjugation with various Ag payloads. We tested T cell activation using VHHs that target distinct APC populations; anti-MHCII adducts elicited strong activation of CD4+ T cells, whereas anti-CD11b showed CD8+ T cell activation superior to targeting via MHCII and CD36. Differences in Ag presentation among constructs were unrelated to dendritic cell subtype or routing to acidic compartments. When coupled to antigenic payloads, anti-MHCII VHH primed Ab responses against GFP, ubiquitin, an OVA peptide, and the α-helix of influenza hemagglutinin's stem; the last afforded protection against influenza infection. The versatility of the VHH scaffold and sortase-mediated covalent attachment of Ags suggests their broader application to generate desirable immune responses.


Asunto(s)
Complejo Antígeno-Anticuerpo/metabolismo , Células Dendríticas/fisiología , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Vacunas contra la Influenza/inmunología , Gripe Humana/inmunología , Infecciones por Orthomyxoviridae/inmunología , Anticuerpos de Dominio Único/metabolismo , Animales , Presentación de Antígeno , Linfocitos T CD4-Positivos/fisiología , Linfocitos T CD8-positivos/fisiología , Camélidos del Nuevo Mundo , Células Cultivadas , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Gripe Humana/prevención & control , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/prevención & control , Anticuerpos de Dominio Único/inmunología
9.
Proc Natl Acad Sci U S A ; 112(36): E4975-84, 2015 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-26305940

RESUMEN

Calcium-dependent protein kinases (CDPKs) comprise the major group of Ca2+-regulated kinases in plants and protists. It has long been assumed that CDPKs are activated, like other Ca2+-regulated kinases, by derepression of the kinase domain (KD). However, we found that removal of the autoinhibitory domain from Toxoplasma gondii CDPK1 is not sufficient for kinase activation. From a library of heavy chain-only antibody fragments (VHHs), we isolated an antibody (1B7) that binds TgCDPK1 in a conformation-dependent manner and potently inhibits it. We uncovered the molecular basis for this inhibition by solving the crystal structure of the complex and simulating, through molecular dynamics, the effects of 1B7-kinase interactions. In contrast to other Ca2+-regulated kinases, the regulatory domain of TgCDPK1 plays a dual role, inhibiting or activating the kinase in response to changes in Ca2+ concentrations. We propose that the regulatory domain of TgCDPK1 acts as a molecular splint to stabilize the otherwise inactive KD. This dependence on allosteric stabilization reveals a novel susceptibility in this important class of parasite enzymes.


Asunto(s)
Proteínas Quinasas/química , Estructura Terciaria de Proteína , Proteínas Protozoarias/química , Toxoplasma/enzimología , Regulación Alostérica , Animales , Anticuerpos Antiprotozoarios/química , Anticuerpos Antiprotozoarios/metabolismo , Anticuerpos Antiprotozoarios/farmacología , Biocatálisis/efectos de los fármacos , Western Blotting , Calcio/metabolismo , Camélidos del Nuevo Mundo , Células Cultivadas , Cristalografía por Rayos X , Activación Enzimática/efectos de los fármacos , Humanos , Cadenas Pesadas de Inmunoglobulina/inmunología , Simulación de Dinámica Molecular , Mutación , Fosforilación , Unión Proteica , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/metabolismo , Anticuerpos de Dominio Único/farmacología , Toxoplasma/genética
10.
bioRxiv ; 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-37645775

RESUMEN

Mycobacterium tuberculosis (Mtb) is known to survive within macrophages by compromising the integrity of the phagosomal compartment in which it resides. This activity primarily relies on the ESX-1 secretion system, predominantly involving the protein duo ESAT-6 and CFP-10. CFP-10 likely acts as a chaperone, while ESAT-6 likely disrupts phagosomal membrane stability via a largely unknown mechanism. we employ a series of biochemical analyses, protein modeling techniques, and a novel ESAT-6-specific nanobody to gain insight into the ESAT-6's mode of action. First, we measure the binding kinetics of the tight 1:1 complex formed by ESAT-6 and CFP-10 at neutral pH. Subsequently, we demonstrate a rapid self-association of ESAT-6 into large complexes under acidic conditions, leading to the identification of a stable tetrameric ESAT-6 species. Using molecular dynamics simulations, we pinpoint the most probable interaction interface. Furthermore, we show that cytoplasmic expression of an anti-ESAT-6 nanobody blocks Mtb replication, thereby underlining the pivotal role of ESAT-6 in intracellular survival. Together, these data suggest that ESAT-6 acts by a pH dependent mechanism to establish two-way communication between the cytoplasm and the Mtb-containing phagosome.

11.
Elife ; 122024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38805257

RESUMEN

Mycobacterium tuberculosis (Mtb) is known to survive within macrophages by compromising the integrity of the phagosomal compartment in which it resides. This activity primarily relies on the ESX-1 secretion system, predominantly involving the protein duo ESAT-6 and CFP-10. CFP-10 likely acts as a chaperone, while ESAT-6 likely disrupts phagosomal membrane stability via a largely unknown mechanism. we employ a series of biochemical analyses, protein modeling techniques, and a novel ESAT-6-specific nanobody to gain insight into the ESAT-6's mode of action. First, we measure the binding kinetics of the tight 1:1 complex formed by ESAT-6 and CFP-10 at neutral pH. Subsequently, we demonstrate a rapid self-association of ESAT-6 into large complexes under acidic conditions, leading to the identification of a stable tetrameric ESAT-6 species. Using molecular dynamics simulations, we pinpoint the most probable interaction interface. Furthermore, we show that cytoplasmic expression of an anti-ESAT-6 nanobody blocks Mtb replication, thereby underlining the pivotal role of ESAT-6 in intracellular survival. Together, these data suggest that ESAT-6 acts by a pH-dependent mechanism to establish two-way communication between the cytoplasm and the Mtb-containing phagosome.


Asunto(s)
Antígenos Bacterianos , Proteínas Bacterianas , Macrófagos , Mycobacterium tuberculosis , Fagosomas , Anticuerpos de Dominio Único , Humanos , Antígenos Bacterianos/metabolismo , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/metabolismo , Concentración de Iones de Hidrógeno , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Simulación de Dinámica Molecular , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/metabolismo , Fagosomas/metabolismo , Anticuerpos de Dominio Único/metabolismo
12.
J Healthc Qual ; 43(6): 355-364, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34267169

RESUMEN

ABSTRACT: Quality measurement across healthcare is undertaken with a goal of improving care and outcomes for patients; however, the relationship between quality measurement and patient outcomes remains largely untested, particularly in inpatient behavioral health. Using a retrospective quantitative design, we assessed 142 behavioral health organizations' quality data submitted to the Hospital-Based Inpatient Psychiatric Services and Inpatient Psychiatric Facility Quality Reporting programs from 2017 to 2018 and tested relationships between compliance on 16 quality measures and symptom improvement on patient self-report outcomes (SROs) at the facility level. Performance on many quality measures was negatively skewed (at least four have almost no room for improvement on average), and there was high interrelatedness between most quality measures. Nine of the assessed measures correlated with patient SROs but not in clear groupings. Findings indicate that an underlying organizational construct may be driving compliance rates on quality measures, but the measures are not linked to treatment outcomes as expected. We encourage an expansion of the current framework of behavioral health quality measurement beyond process and organization and suggest the addition of patient outcomes such as SROs as quality measures to directly assess patient improvement.


Asunto(s)
Atención a la Salud , Pacientes Internos , Humanos , Estudios Retrospectivos
13.
Nat Commun ; 11(1): 6179, 2020 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-33268786

RESUMEN

Nuclear pore complexes (NPCs) are the main conduits for molecular exchange across the nuclear envelope. The NPC is a modular assembly of ~500 individual proteins, called nucleoporins or nups. Most scaffolding nups are organized in two multimeric subcomplexes, the Nup84 or Y complex and the Nic96 or inner ring complex. Working in S. cerevisiae, and to study the assembly of these two essential subcomplexes, we here develop a set of twelve nanobodies that recognize seven constituent nucleoporins of the Y and Nic96 complexes. These nanobodies all bind specifically and with high affinity. We present structures of several nup-nanobody complexes, revealing their binding sites. Additionally, constitutive expression of the nanobody suite in S. cerevisiae detect accessible and obstructed surfaces of the Y complex and Nic96 within the NPC. Overall, this suite of nanobodies provides a unique and versatile toolkit for the study of the NPC.


Asunto(s)
Proteínas de Complejo Poro Nuclear/química , Poro Nuclear/ultraestructura , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/ultraestructura , Anticuerpos de Dominio Único/química , Secuencia de Aminoácidos , Animales , Afinidad de Anticuerpos , Especificidad de Anticuerpos , Sitios de Unión , Camélidos del Nuevo Mundo , Clonación Molecular , Cristalografía por Rayos X , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Cinética , Modelos Moleculares , Poro Nuclear/química , Poro Nuclear/metabolismo , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Complejo Poro Nuclear/metabolismo , Biblioteca de Péptidos , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Anticuerpos de Dominio Único/genética , Anticuerpos de Dominio Único/aislamiento & purificación , Anticuerpos de Dominio Único/metabolismo
14.
Cancer Immunol Res ; 8(4): 518-529, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32019780

RESUMEN

Chimeric antigen receptor (CAR) T-cell therapy is effective in the treatment of cancers of hematopoietic origin. In the immunosuppressive solid tumor environment, CAR T cells encounter obstacles that compromise their efficacy. We developed a strategy to address these barriers by having CAR T cells secrete single-domain antibody fragments [variable heavy domain of heavy chain antibodies (VHH) or nanobodies] that can modify the intratumoral immune landscape and thus support CAR T-cell function in immunocompetent animals. VHHs are small in size and able to avoid domain swapping when multiple nanobodies are expressed simultaneously-features that can endow CAR T cells with desirable properties. The secretion of an anti-CD47 VHH by CAR T cells improves engagement of the innate immune system, enables epitope spreading, and can enhance the antitumor response. CAR T cells that secrete anti-PD-L1 or anti-CTLA-4 nanobodies show improved persistence and demonstrate the versatility of this approach. Furthermore, local delivery of secreted anti-CD47 VHH-Fc fusions by CAR T cells at the tumor site limits their systemic toxicity. CAR T cells can be further engineered to simultaneously secrete multiple modalities, allowing for even greater tailoring of the antitumor immune response.


Asunto(s)
Antígeno CD47/antagonistas & inhibidores , Linfocitos T CD8-positivos/inmunología , Inmunoterapia Adoptiva/métodos , Melanoma Experimental/inmunología , Receptores Quiméricos de Antígenos/inmunología , Proteínas Recombinantes/genética , Anticuerpos de Dominio Único/inmunología , Animales , Línea Celular Tumoral , Granzimas/antagonistas & inhibidores , Melanoma Experimental/genética , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C57BL , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptores Quiméricos de Antígenos/genética , Proteínas Recombinantes/metabolismo , Anticuerpos de Dominio Único/genética , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/metabolismo , Microambiente Tumoral
15.
Open Biol ; 10(2): 190235, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32019478

RESUMEN

Cancer-specific mutations can lead to peptides of unique sequence presented on MHC class I to CD8 T cells. These neoantigens can be potent tumour-rejection antigens, appear to be the driving force behind responsiveness to anti-CTLA-4 and anti-PD1/L1-based therapies and have been used to develop personalized vaccines. The platform for delivering neoantigen-based vaccines has varied, and further optimization of both platform and adjuvant will be necessary to achieve scalable vaccine products that are therapeutically effective at a reasonable cost. Here, we developed a platform for testing potential CD8 T cell tumour vaccine candidates. We used a high-affinity alpaca-derived VHH against MHC class II to deliver peptides to professional antigen-presenting cells. We show in vitro and in vivo that peptides derived from the model antigen ovalbumin are better able to activate naive ovalbumin-specific CD8 T cells when conjugated to an MHC class II-specific VHH when compared with an irrelevant control VHH. We then used the VHH-peptide platform to evaluate a panel of candidate neoantigens in vivo in a mouse model of pancreatic cancer. None of the candidate neoantigens tested led to protection from tumour challenge; however, we were able to show vaccine-induced CD8 T cell responses to a melanoma self-antigen that was augmented by combination therapy with the synthetic cytokine mimetic Neo2/15.


Asunto(s)
Antígenos de Histocompatibilidad Clase II/inmunología , Interleucina-2/administración & dosificación , Melanoma/tratamiento farmacológico , Péptidos/administración & dosificación , Anticuerpos de Dominio Único/metabolismo , Animales , Antígenos de Neoplasias/inmunología , Autoantígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Camélidos del Nuevo Mundo/inmunología , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Sinergismo Farmacológico , Interleucina-2/inmunología , Melanoma/inmunología , Ratones , Péptidos/inmunología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/inmunología , Vacunas de Subunidad , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Elife ; 72018 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-29882741

RESUMEN

Human cytomegalovirus has hijacked and evolved a human G-protein-coupled receptor into US28, which functions as a promiscuous chemokine 'sink' to facilitate evasion of host immune responses. To probe the molecular basis of US28's unique ligand cross-reactivity, we deep-sequenced CX3CL1 chemokine libraries selected on 'molecular casts' of the US28 active-state and find that US28 can engage thousands of distinct chemokine sequences, many of which elicit diverse signaling outcomes. The structure of a G-protein-biased CX3CL1-variant in complex with US28 revealed an entirely unique chemokine amino terminal peptide conformation and remodeled constellation of receptor-ligand interactions. Receptor signaling, however, is remarkably robust to mutational disruption of these interactions. Thus, US28 accommodates and functionally discriminates amongst highly degenerate chemokine sequences by sensing the steric bulk of the ligands, which distort both receptor extracellular loops and the walls of the ligand binding pocket to varying degrees, rather than requiring sequence-specific bonding chemistries for recognition and signaling.


Asunto(s)
Quimiocina CX3CL1/química , Receptores de Quimiocina/química , Receptores Acoplados a Proteínas G/química , Transducción de Señal , Proteínas Virales/química , Animales , Quimiocina CX3CL1/metabolismo , Quimiocina CX3CL1/farmacología , Citomegalovirus/genética , Citomegalovirus/metabolismo , Proteínas de Unión al GTP/química , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Células HEK293 , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Ligandos , Modelos Moleculares , Mutación , Unión Proteica , Conformación Proteica , Receptores de Quimiocina/agonistas , Receptores de Quimiocina/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Virales/agonistas , Proteínas Virales/metabolismo
17.
Cancer Immunol Res ; 6(4): 389-401, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29459478

RESUMEN

Cytokine-based therapies for cancer have not achieved widespread clinical success because of inherent toxicities. Treatment for pancreatic cancer is limited by the dense stroma that surrounds tumors and by an immunosuppressive tumor microenvironment. To overcome these barriers, we developed constructs of single-domain antibodies (VHHs) against PD-L1 fused with IL-2 and IFNγ. Targeting cytokine delivery in this manner reduced pancreatic tumor burden by 50%, whereas cytokines fused to an irrelevant VHH, or blockade of PD-L1 alone, showed little effect. Targeted delivery of IL-2 increased the number of intratumoral CD8+ T cells, whereas IFNγ reduced the number of CD11b+ cells and skewed intratumoral macrophages toward the display of M1-like characteristics. Imaging of fluorescent VHH-IFNγ constructs, as well as transcriptional profiling, demonstrated targeting of IFNγ to the tumor microenvironment. Many tumors and tumor-infiltrating myeloid cells express PD-L1, rendering them potentially susceptible to this form of targeted immunotherapy. Cancer Immunol Res; 6(4); 389-401. ©2018 AACR.


Asunto(s)
Antígeno B7-H1/metabolismo , Citocinas/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Anticuerpos de Dominio Único/farmacología , Microambiente Tumoral , Animales , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Citocinas/antagonistas & inhibidores , Citocinas/genética , Modelos Animales de Enfermedad , Humanos , Melanoma Experimental , Ratones , Terapia Molecular Dirigida , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/terapia , Anticuerpos de Dominio Único/uso terapéutico , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
18.
PLoS One ; 12(12): e0189068, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29200433

RESUMEN

Sortase A, a calcium-dependent transpeptidase derived from Staphylococcus aureus, is used in a broad range of applications, such as the conjugation of fluorescent dyes and other moieties to proteins or to the surface of eukaryotic cells. In vivo and cell-based applications of sortase have been somewhat limited by the large range of calcium concentrations, as well as by the often transient nature of protein-protein interactions in living systems. In order to use sortase A for cell labeling applications, we generated a new sortase A variant by combining multiple mutations to yield an enzyme that was both calcium-independent and highly active. This variant has enhanced activity for both N- and C-terminal labeling, as well as for cell surface modification under physiological conditions.


Asunto(s)
Aminoaciltransferasas/genética , Proteínas Bacterianas/genética , Calcio/metabolismo , Cisteína Endopeptidasas/genética , Peptidil Transferasas/genética , Coloración y Etiquetado/métodos , Staphylococcus aureus/genética , Aminoaciltransferasas/metabolismo , Proteínas Bacterianas/metabolismo , Membrana Celular/química , Cisteína Endopeptidasas/metabolismo , Mutación , Peptidil Transferasas/metabolismo , Staphylococcus aureus/enzimología
19.
Nat Commun ; 8(1): 647, 2017 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-28935898

RESUMEN

Programmed death ligand 1 (PD-L1) is expressed on a number of immune and cancer cells, where it can downregulate antitumor immune responses. Its expression has been linked to metabolic changes in these cells. Here we develop a radiolabeled camelid single-domain antibody (anti-PD-L1 VHH) to track PD-L1 expression by immuno-positron emission tomography (PET). PET-CT imaging shows a robust and specific PD-L1 signal in brown adipose tissue (BAT). We confirm expression of PD-L1 on brown adipocytes and demonstrate that signal intensity does not change in response to cold exposure or ß-adrenergic activation. This is the first robust method of visualizing murine brown fat independent of its activation state.Current approaches to visualise brown adipose tissue (BAT) rely primarily on markers that reflect its metabolic activity. Here, the authors show that PD-L1 is expressed on brown adipocytes, does not change upon BAT activation, and that BAT volume in mice can be measured by PET-CT with a radiolabeled anti-PD-L1 antibody.


Asunto(s)
Adipocitos Marrones/metabolismo , Tejido Adiposo Pardo/metabolismo , Antígeno B7-H1/análisis , Biomarcadores/análisis , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/diagnóstico por imagen , Animales , Antígeno B7-H1/genética , Antígeno B7-H1/inmunología , Camélidos del Nuevo Mundo/inmunología , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Reproducibilidad de los Resultados
20.
J Exp Med ; 214(8): 2243-2255, 2017 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-28666979

RESUMEN

Immunotherapy using checkpoint-blocking antibodies against targets such as CTLA-4 and PD-1 can cure melanoma and non-small cell lung cancer in a subset of patients. The presence of CD8 T cells in the tumor correlates with improved survival. We show that immuno-positron emission tomography (immuno-PET) can visualize tumors by detecting infiltrating lymphocytes and, through longitudinal observation of individual animals, distinguish responding tumors from those that do not respond to therapy. We used 89Zr-labeled PEGylated single-domain antibody fragments (VHHs) specific for CD8 to track the presence of intratumoral CD8+ T cells in the immunotherapy-susceptible B16 melanoma model in response to checkpoint blockade. A 89Zr-labeled PEGylated anti-CD8 VHH detected thymus and secondary lymphoid structures as well as intratumoral CD8 T cells. Animals that responded to CTLA-4 therapy showed a homogeneous distribution of the anti-CD8 PET signal throughout the tumor, whereas more heterogeneous infiltration of CD8 T cells correlated with faster tumor growth and worse responses. To support the validity of these observations, we used two different transplantable breast cancer models, yielding results that conformed with predictions based on the antimelanoma response. It may thus be possible to use immuno-PET and monitor antitumor immune responses as a prognostic tool to predict patient responses to checkpoint therapies.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Antígeno CTLA-4/antagonistas & inhibidores , Neoplasias Mamarias Experimentales/terapia , Animales , Antígeno CTLA-4/fisiología , Femenino , Fragmentos de Inmunoglobulinas/inmunología , Fragmentos de Inmunoglobulinas/uso terapéutico , Inmunoterapia/métodos , Neoplasias Mamarias Experimentales/diagnóstico por imagen , Neoplasias Mamarias Experimentales/inmunología , Ratones , Trasplante de Neoplasias , Tomografía de Emisión de Positrones/métodos , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA