Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Curr Opin Hematol ; 29(2): 74-83, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35013048

RESUMEN

PURPOSE OF REVIEW: Treatment outcome of relapsed or refractory AML patients remains dismal and new treatment options are needed. Adoptive cell therapy using CAR-T cells is a potentially interesting approach in this. RECENT FINDINGS: Several potentially interesting AML targets are being investigated with CAR-T therapy with over 60 clinical trials listed on clinicaltrials.gov. The first clinical data are only just emerging with mixed results, once more proving that further research is needed. SUMMARY: Adoptive cell therapy using chimeric antigen receptor T cells is being investigated in AML through many clinical trials. So far, no AML-specific antigen has been identified, requiring additional strategies to mitigate on-target off-tumor toxicity and to increase efficacy. Focus point is to acquire control over the CAR T cells once administered. Strategies to do so include biodegradable CARs, inducible CARs, suicide-switch containing CARs and two-component modular CARs. Limited and mixed results are available, confirming the risk of lasting toxicity for nonswitchable CARs. Initial results of modular CARs suggest toxicity can be mitigated whilst maintaining CAR activity by the use of modular CAR concepts that allows for 'ON' and 'OFF' switching.


Asunto(s)
Leucemia Mieloide Aguda , Receptores Quiméricos de Antígenos , Tratamiento Basado en Trasplante de Células y Tejidos , Humanos , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos , Leucemia Mieloide Aguda/patología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/uso terapéutico
2.
J Immunol ; 202(6): 1735-1746, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30728213

RESUMEN

Long-term survival of adoptively transferred chimeric Ag receptor (CAR) T cells is often limited. Transplantation of hematopoietic stem cells (HSCs) transduced to express CARs could help to overcome this problem as CAR-armed HSCs can continuously deliver CAR+ multicell lineages (e.g., T cells, NK cells). In dependence on the CAR construct, a variable extent of tonic signaling in CAR T cells was reported; thus, effects of CAR-mediated tonic signaling on the hematopoiesis of CAR-armed HSCs is unclear. To assess the effects of tonic signaling, two CAR constructs were established and analyzed 1) a signaling CAR inducing a solid Ag-independent tonic signaling termed CAR-28/ζ and 2) a nonstimulating control CAR construct lacking intracellular signaling domains termed CAR-Stop. Bone marrow cells from immunocompetent mice were isolated, purified for HSC-containing Lin-cKit+ cells or the Lin-cKit+ Sca-1+ subpopulation (Lin-Sca-1+cKit+), and transduced with both CAR constructs. Subsequently, modified bone marrow cells were transferred into irradiated mice, in which they successfully engrafted and differentiated into hematopoietic progenitors. HSCs expressing the CAR-Stop sustained normal hematopoiesis. In contrast, expression of the CAR-28/ζ led to elimination of mature CAR+ T and B cells, suggesting that the CAR-mediated tonic signaling mimics autorecognition via the newly recombined immune receptors in the developing lymphocytes.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Activación de Linfocitos/fisiología , Linfopoyesis/fisiología , Receptores Quiméricos de Antígenos/metabolismo , Transducción de Señal/fisiología , Traslado Adoptivo , Animales , Diferenciación Celular/fisiología , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Ratones , Ratones Endogámicos C57BL
3.
Cancer Immunol Immunother ; 68(9): 1401-1415, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31414180

RESUMEN

Although CAR T-cell therapy has demonstrated tremendous clinical efficacy especially in hematological malignancies, severe treatment-associated toxicities still compromise the widespread application of this innovative technology. Therefore, developing novel approaches to abrogate CAR T-cell-mediated side effects is of great relevance. Several promising strategies pursue the selective antibody-based depletion of adoptively transferred T cells via elimination markers. However, given the limited half-life and tissue penetration, dependence on the patients' immune system and on-target/off-side effects of proposed monoclonal antibodies, we sought to exploit αCAR-engineered T cells to efficiently eliminate CAR T cells. For comprehensive and specific recognition, a small peptide epitope (E-tag) was incorporated into the extracellular spacer region of CAR constructs. We provide first proof-of-concept for targeting this epitope by αE-tag CAR T cells, allowing an effective killing of autologous E-tagged CAR T cells both in vitro and in vivo whilst sparing cells lacking the E-tag. In addition to CAR T-cell cytotoxicity, the αE-tag-specific T cells can be empowered with cancer-fighting ability in case of relapse, hence, have versatile utility. Our proposed methodology can most probably be implemented in CAR T-cell therapies regardless of the targeted tumor antigen aiding in improving overall safety and survival control of highly potent gene-modified cells.


Asunto(s)
Epítopos de Linfocito T/genética , Inmunoterapia Adoptiva/métodos , Fragmentos de Péptidos/genética , Neoplasias de la Próstata/terapia , Receptores de Antígenos de Linfocitos T/genética , Receptores Quiméricos de Antígenos/genética , Linfocitos T Citotóxicos/inmunología , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Autoantígenos/inmunología , Citotoxicidad Inmunológica , Epítopos de Linfocito T/inmunología , Ingeniería Genética , Humanos , Masculino , Ratones , Recurrencia Local de Neoplasia , Células PC-3 , Neoplasias de la Próstata/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
4.
J Autoimmun ; 90: 116-131, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29503042

RESUMEN

As regulatory T cells (Tregs) play a fundamental role in immune homeostasis their adoptive transfer emerged as a promising treatment strategy for inflammation-related diseases. Preclinical animal models underline the superiority of antigen-specific Tregs compared to polyclonal cells. Here, we applied a modular chimeric antigen receptor (CAR) technology called UniCAR for generation of antigen-specific human Tregs. In contrast to conventional CARs, UniCAR-endowed Tregs are indirectly linked to their target cells via a separate targeting module (TM). Thus, transduced Tregs can be applied universally as their antigen-specificity is easily adjusted by TM exchange. Activation of UniCAR-engrafted Tregs occurred in strict dependence on the TM, facilitating a precise control over Treg activity. In order to augment efficacy and safety, different intracellular signaling domains were tested. Both 4-1BB (CD137) and CD28 costimulation induced strong suppressive function of genetically modified Tregs. However, in light of safety issues, UniCARs comprising a CD137-CD3ζ signaling domain emerged as constructs of choice for a clinical application of redirected Tregs. In that regard, Tregs isolated from patients suffering from autoimmune or inflammatory diseases were, for the first time, successfully engineered with UniCAR 137/ζ and efficiently suppressed patient-derived effector cells. Overall, the UniCAR platform represents a promising approach to improve Treg-based immunotherapies for tolerance induction.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Receptores de Antígenos de Linfocitos T/genética , Receptores Quiméricos de Antígenos/genética , Linfocitos T Reguladores/fisiología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Traslado Adoptivo , Animales , Células Cultivadas , Células HEK293 , Humanos , Ratones , Células 3T3 NIH , Receptores de Antígenos/genética , Especificidad del Receptor de Antígeno de Linfocitos T
6.
J Immunol ; 194(7): 3201-12, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25740942

RESUMEN

NK cells are emerging as new effectors for immunotherapy of cancer. In particular, the genetic engraftment of chimeric Ag receptors (CARs) in NK cells is a promising strategy to redirect NK cells to otherwise NK cell-resistant tumor cells. On the basis of DNAX-activation protein 12 (DAP12), a signaling adaptor molecule involved in signal transduction of activating NK cell receptors, we generated a new type of CAR targeting the prostate stem cell Ag (PSCA). We demonstrate in this article that this CAR, designated anti-PSCA-DAP12, consisting of DAP12 fused to the anti-PSCA single-chain Ab fragment scFv(AM1) confers improved cytotoxicity to the NK cell line YTS against PSCA-positive tumor cells when compared with a CAR containing the CD3ζ signaling chain. Further analyses revealed phosphorylation of the DAP12-associated ZAP-70 kinase and IFN-γ release of CAR-engineered cells after contact with PSCA-positive target cells. YTS cells modified with DAP12 alone or with a CAR bearing a phosphorylation-defective ITAM were not activated. Notably, infused YTS cells armed with anti-PSCA-DAP12 caused delayed tumor xenograft growth and resulted in complete tumor eradication in a significant fraction of treated mice. The feasibility of the DAP12-based CAR was further tested in human primary NK cells and confers specific cytotoxicity against KIR/HLA-matched PSCA-positive tumor cells, which was further enhanced by KIR-HLA mismatches. We conclude that NK cells engineered with DAP12-based CARs are a promising tool for adoptive tumor immunotherapy.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Proteínas de la Membrana/genética , Neoplasias/genética , Neoplasias/inmunología , Receptores de Células Asesinas Naturales/genética , Proteínas Recombinantes de Fusión , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Complejo CD3/genética , Complejo CD3/inmunología , Línea Celular , Línea Celular Tumoral , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Femenino , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/inmunología , Vectores Genéticos/genética , Humanos , Inmunofenotipificación , Inmunoterapia , Inmunoterapia Adoptiva , Interferón gamma/biosíntesis , Ratones , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/mortalidad , Neoplasias/patología , Neoplasias/terapia , Fenotipo , Fosforilación , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína Tirosina Quinasa ZAP-70/metabolismo
7.
Hepatology ; 60(4): 1196-210, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24845056

RESUMEN

UNLABELLED: The low-grade inflammatory state present in obesity contributes to obesity-related metabolic dysregulation, including nonalcoholic steatohepatitis (NASH) and insulin resistance. Intercellular interactions between immune cells or between immune cells and hepatic parenchymal cells contribute to the exacerbation of liver inflammation and steatosis in obesity. The costimulatory molecules, B7.1 and B7.2, are important regulators of cell-cell interactions in several immune processes; however, the role of B7 costimulation in obesity-related liver inflammation is unknown. Here, diet-induced obesity (DIO) studies in mice with genetic inactivation of both B7.1 and B7.2 (double knockout; DKO) revealed aggravated obesity-related metabolic dysregulation, reduced insulin signalling in the liver and adipose tissue (AT), glucose intolerance, and enhanced progression to steatohepatitis resulting from B7.1/B7.2 double deficiency. The metabolic phenotype of B7.1/B7.2 double deficiency upon DIO was accompanied by increased hepatic and AT inflammation, associated with largely reduced numbers of regulatory T cells (Tregs) in these organs. In order to assess the role of B7 costimulation in DIO in a non-Treg-lacking environment, we performed antibody (Ab)-mediated inhibition of B7 molecules in wild-type mice in DIO. Antibody-blockade of both B7.1 and B7.2 improved the metabolic phenotype of DIO mice, which was linked to amelioration of hepatic steatosis and reduced inflammation in liver and AT. CONCLUSION: Our study demonstrates a dual role of B7 costimulation in the course of obesity-related sequelae, particularly NASH. The genetic inactivation of B7.1/B7.2 deteriorates obesity-related liver steatosis and metabolic dysregulation, likely a result of the intrinsic absence of Tregs in these mice, rendering DKO mice a novel murine model of NASH. In contrast, inhibition of B7 costimulation under conditions where Tregs are present may provide a novel therapeutic approach for obesity-related metabolic dysregulation and, especially, NASH.


Asunto(s)
Antígenos B7/fisiología , Síndrome Metabólico/fisiopatología , Enfermedad del Hígado Graso no Alcohólico/fisiopatología , Obesidad/fisiopatología , Animales , Antígenos B7/deficiencia , Antígenos B7/genética , Comunicación Celular/fisiología , Modelos Animales de Enfermedad , Hígado/patología , Masculino , Ratones , Ratones Noqueados , Fenotipo , Linfocitos T Reguladores/patología
8.
Prostate ; 74(13): 1335-46, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25053443

RESUMEN

BACKGROUND: Recently, we described a novel modular platform technology in which T cell-recruitment and tumor-targeting domains of conventional bispecific antibodies are split to independent components, a universal effector module (EM) and replaceable monospecific/monovalent target modules (TMs) that form highly efficient T cell-retargeting complexes. Theoretically, our unique strategy should allow us to simultaneously retarget T cells to different tumor antigens by combining the EM with two or more different monovalent/monospecific TMs or even with bivalent/bispecific TMs, thereby overcoming limitations of a monospecific treatment such as the selection of target-negative tumor escape variants. METHODS: In order to advance our recently introduced prostate stem cell antigen (PSCA)-specific modular system for a dual-targeting of prostate cancer cells, two additional TMs were constructed: a monovalent/monospecific TM directed against the prostate-specific membrane antigen (PSMA) and a bivalent/bispecific TM (bsTM) with specificity for PSMA and PSCA. The functionality of the novel dual-targeting strategies was analyzed by performing T cell activation and chromium release assays. RESULTS: Similar to the PSCA-specific modular system, the novel PSMA-specific modular system mediates an efficient target-dependent and -specific tumor cell lysis at low E:T ratios and picomolar Ab concentrations. Moreover, by combination of the EM with either the bispecific TM directed to PSMA and PSCA or both monospecifc TMs directed to either PSCA or PSMA, dual-specific targeting complexes were formed which allowed us to kill potential escape variants expressing only one or the other target antigen. CONCLUSIONS: Overall, the novel modular system represents a promising tool for multiple tumor targeting.


Asunto(s)
Antígenos de Neoplasias/inmunología , Antígenos de Superficie/inmunología , Glutamato Carboxipeptidasa II/inmunología , Proteínas de Neoplasias/inmunología , Neoplasias de la Próstata/terapia , Linfocitos T/inmunología , Línea Celular Tumoral , Proteínas Ligadas a GPI/inmunología , Células HEK293 , Humanos , Inmunoterapia , Masculino , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/patología , Linfocitos T/patología
9.
Prostate ; 74(13): 1347-58, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25053504

RESUMEN

BACKGROUND: There is still a need for new therapeutic options against prostate cancer. Conventional single-chain bispecific antibodies (bsAbs), that directly cross-link T cells and tumor cells, hold great potential for efficient tumor treatment. However, rapid development of novel bsAbs is hampered by laborious optimization to improve their efficacy and reduce potential side effects. To accelerate the development of a novel antibody tool for the redirection of T cells to different tumor-associated antigens, we recently introduced a modular targeting system. METHODS: We here describe a novel modular system for treatment of prostate cancer by retargeting of T cells to the prostate stem cell antigen (PSCA). Functionality of the novel PSCA-specific modular system was investigated in vitro by T cell activation and chromium release assays as well as in immunodeficient mice. RESULTS: Similar to a conventional bsAb CD3-PSCA, the novel PSCA-specific modular system induces activation of both CD4+ and CD8+ T cells leading to secretion of pro-inflammatory cytokines and highly efficient target-specific tumor cell lysis. The novel TM was ready-to-use from the time point of construction and functional at low E:T ratios and picomolar concentrations without further optimization. In addition, the PSCA-specific modular system delays outgrowth of s.c. tumors in mice comparable to bsAb CD3-PSCA. CONCLUSIONS: We have developed a novel PSCA-specific modular system which triggers an efficient T cell-mediated killing of PSCA+ tumor cells in vitro and in vivo. The new Ab-based targeting strategy can functionally replace conventional bsAbs and allows a flexible redirection of T cells to different tumor-associated antigens.


Asunto(s)
Antígenos de Neoplasias/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Proteínas de Neoplasias/inmunología , Neoplasias de la Próstata/terapia , Animales , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/patología , Línea Celular Tumoral , Proteínas Ligadas a GPI/inmunología , Humanos , Inmunoterapia , Masculino , Ratones , Próstata/inmunología , Próstata/patología , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/patología
10.
J Immunol ; 189(6): 3249-59, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22875801

RESUMEN

Prostate cancer is the most common noncutaneous malignancy in men. The prostate stem cell Ag (PSCA) is a promising target for immunotherapy of advanced disease. Based on a novel mAb directed to PSCA, we established and compared a series of murine and humanized anti-CD3-anti-PSCA single-chain bispecific Abs. Their capability to redirect T cells for killing of tumor cells was analyzed. During these studies, we identified a novel bispecific humanized Ab that efficiently retargets T cells to tumor cells in a strictly Ag-dependent manner and at femtomolar concentrations. T cell activation, cytokine release, and lysis of target cells depend on a cross-linkage of redirected T cells with tumor cells, whereas binding of the anti-CD3 domain alone does not lead to an activation or cytokine release. Interestingly, both CD8+ and CD4+ T cells are activated in parallel and can efficiently mediate the lysis of tumor cells. However, the onset of killing via CD4+ T cells is delayed. Furthermore, redirecting T cells via the novel humanized bispecific Abs results in a delay of tumor growth in xenografted nude mice.


Asunto(s)
Anticuerpos Biespecíficos/fisiología , Anticuerpos Monoclonales Humanizados/fisiología , Antígenos de Neoplasias/biosíntesis , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Proteínas de Neoplasias/biosíntesis , Neoplasias de la Próstata/inmunología , Células Madre/inmunología , Ensayo de Tumor de Célula Madre , Antígenos de Neoplasias/inmunología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/patología , Muerte Celular/inmunología , Epítopos de Linfocito T/inmunología , Proteínas Ligadas a GPI/antagonistas & inhibidores , Proteínas Ligadas a GPI/biosíntesis , Proteínas Ligadas a GPI/inmunología , Humanos , Masculino , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/inmunología , Neoplasias de la Próstata/patología , Células Madre/patología , Ensayo de Tumor de Célula Madre/métodos
11.
J Immunol ; 188(3): 1551-8, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22184723

RESUMEN

Bispecific Abs hold great potential for immunotherapy of malignant diseases. Because the first components of this new drug class are now entering clinical trials, all aspects of their mode of action should be well understood. Several studies proved that CD8(+) and CD4(+) effector T cells can be successfully redirected and activated against tumor cells by bispecific Abs both in vitro and in vivo. To our knowledge, this study provides the first evidence that bispecific Abs can also redirect and activate regulatory T cells against a surface Ag, independently of their TCR specificity. After cross-linking, via a bispecific Ab, redirected regulatory T cells upregulate the activation markers CD69 and CD25, as well as regulatory T cell-associated markers, like CTLA-4 and FOXP3. The activated regulatory T cells secrete the immunosuppressive cytokine IL-10, but, in contrast to CD8(+) and CD4(+) effector T cells, almost no inflammatory cytokines. In addition, the redirected regulatory T cells are able to suppress effector functions of activated autologous CD4(+) T cells both in vitro and in vivo. Therefore, the potential risk for activation of regulatory T cells should be taken into consideration when bispecific Abs are applied for the treatment of malignant diseases. In contrast, an Ag/tissue-specific redirection of regulatory T cells with bispecific Abs holds great potential for the treatment of autoimmune diseases and graft rejection.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Terapia Molecular Dirigida/métodos , Linfocitos T Reguladores/efectos de los fármacos , Anticuerpos Biespecíficos/farmacología , Antígenos de Superficie/efectos de los fármacos , Línea Celular , Humanos , Interleucina-10 , Activación de Linfocitos
12.
J Autoimmun ; 42: 105-16, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23352111

RESUMEN

The nuclear autoantigen La can be detected on the surface of dying cells. Here we present an assay which enables us to show that La protein is not limited to the surface of dying cells but will be released upon stress-induced cell death. As released La protein tightly binds to the surface of neighboring intact cells we asked the question whether or not La protein could serve as a stress-inducible target e.g. for redirecting of regulatory T cells (Tregs) into damaged tissues to downregulate an immune response. In order to provide first proof of concept we developed a novel fully humanized single-chain bispecific antibody (bsAb) which on the one hand is directed to the La antigen and on the other hand to the CD3 complex of T cells. A cross-linkage of Tregs with La-decorated target cells mediated by this bsAb resulted indeed in the activation of the Tregs in a target-dependent manner. Moreover, such bsAb activated Tregs displayed a potent suppressive capacity and negatively influenced proliferation, expansion and cytokine production of autologous CD4(+) and CD8(+) Teff cells.


Asunto(s)
Autoantígenos/inmunología , Complejo CD3/inmunología , Terapia de Inmunosupresión , Proteínas de la Membrana/inmunología , Ribonucleoproteínas/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Células 3T3 , Animales , Anticuerpos Monoclonales Humanizados/inmunología , Autoantígenos/genética , Complejo CD3/genética , Muerte Celular/efectos de los fármacos , Muerte Celular/inmunología , Células HEK293 , Células HeLa , Humanos , Activación de Linfocitos/efectos de los fármacos , Proteínas de la Membrana/genética , Ratones , Receptor Cross-Talk/inmunología , Ribonucleoproteínas/genética , Anticuerpos de Cadena Única/inmunología , Estrés Fisiológico/inmunología , Linfocitos T Reguladores/inmunología , Antígeno SS-B
13.
Anal Biochem ; 423(2): 261-8, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22274538

RESUMEN

There is growing interest in the development of novel single-chain bispecific antibodies for retargeting of immune effector T cells to tumor cells. Until today, functional fusion constructs consisting of a single-chain bispecific antibody and a fluorescent protein were not reported. Such molecules could be useful for an in vivo visualization of this retargeting process. Recently, we established two novel single-chain bispecific antibodies. One is capable of retargeting T cells to CD33, and the other is capable of retargeting T cells to the prostate stem cell antigen (PSCA). CD33 is an attractive immunotarget on the surface of tumor cells from patients with acute myeloid leukemia (AML). The PSCA is a potential target on prostate cancer cells. Flanking the reading frame encoding the green fluorescent protein (GFP) with a recently described novel helical linker element allowed us to establish novel single-chain bispecific fusion antibodies. These fluorescent fusion antibodies were useful to efficiently retarget T cells to the respective tumor cells and visualize the formation of immune synapses between effector and target cells.


Asunto(s)
Anticuerpos Biespecíficos/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Sinapsis Inmunológicas/patología , Microscopía Confocal , Linfocitos T/metabolismo , Anticuerpos Biespecíficos/genética , Anticuerpos Biespecíficos/inmunología , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Antígenos CD/inmunología , Antígenos de Diferenciación Mielomonocítica/inmunología , Complejo CD3/inmunología , Línea Celular Tumoral , Proteínas Fluorescentes Verdes/genética , Células HEK293 , Humanos , Sinapsis Inmunológicas/metabolismo , Antígeno Prostático Específico/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Lectina 3 Similar a Ig de Unión al Ácido Siálico , Linfocitos T/inmunología
14.
Prostate ; 71(9): 998-1011, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21541976

RESUMEN

BACKGROUND: Prostate cancer (PCa) is the most common malignant disease in men. Novel treatment options are needed for patients after development of metastatic, hormone-refractory disease or for those who have failed a local treatment. The prostate stem cell antigen (PSCA) is expressed in >80% of primary PCa samples and bone metastases. Its expression is increased both in androgen-dependent and independent prostate tumors, particularly in carcinomas of high stages and Gleason scores. Therefore, PSCA is an attractive target for immunotherapy of PCa by retargeting of T cells to tumor cells. METHODS: A series of different bispecific antibody formats for retargeting of T cells to tumor cells were described but, only very limited data obtained by side by side comparison of the different antibody formats are available. We established two novel bispecific antibodies in different formats. The functionality of both constructs was analyzed by FACS and chromium release assays. In parallel, the release of pro-inflammatory cytokines was determined by ELISA. RESULTS AND CONCLUSIONS: Irrespective of the underlying antibody format, both novel bispecific antibodies cause an efficient killing of PSCA-positive tumor cells by pre- and non-pre-activated T cells. Killing and release of pro-inflammatory cytokines requires an antigen specific cross-linkage of the T cells with the target cells.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Antígenos de Neoplasias/inmunología , Proteínas de Neoplasias/inmunología , Neoplasias de la Próstata/inmunología , Linfocitos T/inmunología , Anticuerpos Biespecíficos/inmunología , Línea Celular Tumoral , Citocinas/análisis , Pruebas Inmunológicas de Citotoxicidad , Citometría de Flujo , Proteínas Ligadas a GPI/inmunología , Humanos , Inmunización Pasiva/métodos , Masculino , Neoplasias Hormono-Dependientes/inmunología , Neoplasias Hormono-Dependientes/terapia , Neoplasias de la Próstata/terapia , ARN/química , ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
J Biomed Biotechnol ; 2011: 918471, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22190859

RESUMEN

An intact immune system is essential to prevent the development and progression of neoplastic cells in a process termed immune surveillance. During this process the innate and the adaptive immune systems closely cooperate and especially T cells play an important role to detect and eliminate tumor cells. Due to the mechanism of central tolerance the frequency of T cells displaying appropriate arranged tumor-peptide-specific-T-cell receptors is very low and their activation by professional antigen-presenting cells, such as dendritic cells, is frequently hampered by insufficient costimulation resulting in peripheral tolerance. In addition, inhibitory immune circuits can impair an efficient antitumoral response of reactive T cells. It also has been demonstrated that large tumor burden can promote a state of immunosuppression that in turn can facilitate neoplastic progression. Moreover, tumor cells, which mostly are genetically instable, can gain rescue mechanisms which further impair immune surveillance by T cells. Herein, we summarize the data on how tumor cells evade T-cell immune surveillance with the focus on solid tumors and describe approaches to improve anticancer capacity of T cells.


Asunto(s)
Vigilancia Inmunológica , Neoplasias/inmunología , Linfocitos T/inmunología , Escape del Tumor , Animales , Células Dendríticas/inmunología , Humanos , Tolerancia Inmunológica , Proteínas de Neoplasias/inmunología , Neoplasias/patología , Péptidos/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/patología
16.
Oncoimmunology ; 10(1): 1945804, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34290907

RESUMEN

Chimeric antigen receptor T cells (CAR-T) targeting CD19 have achieved significant success in patients with B cell malignancies. To date, implementation of CAR-T in other indications remains challenging due to the lack of truly tumor-specific antigens as well as control of CAR-T activity in patients. CD123 is highly expressed in acute myeloid leukemia (AML) blasts including leukemia-initiating cells making it an attractive immunotherapeutic target. However, CD123 expression in normal hematopoietic progenitor cells and endothelia bears the risk of severe toxicities and may limit CAR-T applications lacking fine-tuned control mechanisms. Therefore, we recently developed a rapidly switchable universal CAR-T platform (UniCAR), in which CAR-T activity depends on the presence of a soluble adapter called targeting module (TM), and confirmed clinical proof-of-concept for targeting CD123 in AML with improved safety. As costimulation via 4-1BB ligand (4-1BBL) can enhance CAR-T expansion, persistence, and effector functions, a novel CD123-specific TM variant (TM123-4-1BBL) comprising trimeric single-chain 4-1BBL was developed for transient costimulation of UniCAR-T cells (UniCAR-T) at the leukemic site in trans. TM123-4-1BBL-directed UniCAR-T efficiently eradicated CD123-positive AML cells in vitro and in a CDX in vivo model. Moreover, additional costimulation via TM123-4-1BBL enabled enhanced expansion and persistence with a modulated UniCAR-T phenotype. In addition, the increased hydrodynamic volume of TM123-4-1BBL prolonged terminal plasma half-life and ensured a high total drug exposure in vivo. In conclusion, expanding the soluble adapter optionality for CD123-directed UniCAR-T maintains the platforms high anti-leukemic efficacy and immediate control mechanism for a flexible, safe, and individualized CAR-T therapy of AML patients.


Asunto(s)
Subunidad alfa del Receptor de Interleucina-3 , Leucemia Mieloide Aguda , Antígenos de Neoplasias , Humanos , Inmunoterapia Adoptiva , Leucemia Mieloide Aguda/tratamiento farmacológico , Linfocitos T
17.
J Biomed Biotechnol ; 2010: 956304, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20467460

RESUMEN

CD4+ and CD8+ T lymphocytes are powerful components of adaptive immunity, which essentially contribute to the elimination of tumors. Due to their cytotoxic capacity, T cells emerged as attractive candidates for specific immunotherapy of cancer. A promising approach is the genetic modification of T cells with chimeric antigen receptors (CARs). First generation CARs consist of a binding moiety specifically recognizing a tumor cell surface antigen and a lymphocyte activating signaling chain. The CAR-mediated recognition induces cytokine production and tumor-directed cytotoxicity of T cells. Second and third generation CARs include signal sequences from various costimulatory molecules resulting in enhanced T-cell persistence and sustained antitumor reaction. Clinical trials revealed that the adoptive transfer of T cells engineered with first generation CARs represents a feasible concept for the induction of clinical responses in some tumor patients. However, further improvement is required, which may be achieved by second or third generation CAR-engrafted T cells.


Asunto(s)
Antígenos de Neoplasias , Inmunoterapia/métodos , Neoplasias/terapia , Ingeniería de Proteínas/métodos , Receptores de Antígenos de Linfocitos T , Proteínas Recombinantes de Fusión/uso terapéutico , Animales , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Humanos , Ratones , Modelos Inmunológicos , Neoplasias/inmunología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal/inmunología
18.
Mol Ther Oncolytics ; 17: 408-420, 2020 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-32462078

RESUMEN

Chimeric antigen receptor T cells (CAR-T) targeting CD19 or B cell maturation antigen (BCMA) are highly effective against B cell malignancies. However, application of CAR-T to less differentially expressed targets remains a challenge due to lack of tumor-specific antigens and CAR-T controllability. CD123, a highly promising leukemia target, is expressed not only by leukemic and leukemia-initiating cells, but also by myeloid, hematopoietic progenitor, and certain endothelial cells. Thus, CAR-T lacking fine-tuned control mechanisms pose a high toxicity risk. To extend the CAR-T target landscape and widen the therapeutic window, we adapted our rapidly switchable universal CAR-T platform (UniCAR) to target CD123. UniCAR-T efficiently eradicated CD123+ leukemia in vitro and in vivo. Activation, cytolytic response, and cytokine release were strictly dependent on the presence of the CD123-specific targeting module (TM123) with comparable efficacy to CD123-specific CAR-T in vitro. We further demonstrated a pre-clinical proof of concept for the safety-switch mechanism using a hematotoxicity mouse model wherein TM123-redirected UniCAR-T showed reversible toxicity toward hematopoietic cells compared to CD123 CAR-T. In conclusion, UniCAR-T maintain full anti-leukemic efficacy, while ensuring rapid controllability to improve safety and versatility of CD123-directed immunotherapy. The safety and efficacy of UniCAR-T in combination with TM123 will now be assessed in a phase I clinical trial (ClinicalTrials.gov: NCT04230265).

19.
Int J Cancer ; 124(2): 358-66, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-18942710

RESUMEN

Dendritic cells (DCs) display an extraordinary capacity to induce T-cell responses providing the opportunity of DC-based cancer vaccination strategies. Additional findings indicate that DCs may also play a crucial role for the activation of natural killer (NK) cells, which are important effectors in innate antitumor immunity. However, studies investigating the interaction between native human DCs and NK cells are limited. Recently, we defined 6-sulfo LacNAc (slan) DCs as a major subpopulation of myeloid human blood DCs, which represent principal producers of the proinflammatory cytokines tumor necrosis factor-alpha and interleukin (IL)-12. Functional data revealed that slanDCs efficiently induce neoantigen-specific CD4+ T cells and activate tumor-reactive cytotoxic T cells. When evaluating the crosstalk between slanDCs and NK cells in this study, we found that lipopolysaccharide (LPS)-activated slanDCs efficiently enhance NK cell CD69 expression and interferon (IFN)-gamma secretion. NK cell-mediated tumor-directed cytotoxicity was significantly improved by slanDCs. NK cell activation induced by slanDCs was critically dependent on IL-12. When investigating the impact of NK cells on the immunostimulatory capacity of slanDCs, we observed that they promote DC maturation. In addition, NK cells strongly enhanced the secretion of immunomodulatory IL-12 and reduced the release of immunosuppressive IL-10 by slanDCs. IFN-gamma and cell-to-cell contact contributed to these effects. Furthermore, data revealed that DC-NK cell crosstalk improves slanDC-mediated differentiation of naïve CD4+ T lymphocytes into IFN-gamma-producing Th1 cells. In conclusion, we demonstrate a reciprocal activating interaction between slanDCs and NK cells, which may play a pivotal role in the regulation of antitumor immunity.


Asunto(s)
Amino Azúcares/metabolismo , Células Dendríticas/metabolismo , Células Asesinas Naturales/metabolismo , Antígenos CD/biosíntesis , Antígenos de Diferenciación de Linfocitos T/biosíntesis , Citocinas/metabolismo , Células Dendríticas/citología , Humanos , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Interleucina-12/metabolismo , Células K562 , Lectinas Tipo C , Leucocitos Mononucleares/citología , Lipopolisacáridos/metabolismo , Activación de Linfocitos , Modelos Biológicos , Factor de Necrosis Tumoral alfa/metabolismo
20.
Mol Ther Nucleic Acids ; 18: 708-726, 2019 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-31726388

RESUMEN

The adaptation of CRISPR/Cas technology for use in mammals has revolutionized genome engineering. In particular with regard to clinical application, efficient expression of Cas9 within a narrow time frame is highly desirable to minimize the accumulation of off-target editing. We developed an effective, aptamer-independent retroviral delivery system for Cas9 mRNAs that takes advantage of a unique foamy virus (FV) capability: the efficient encapsidation and transfer of non-viral RNAs. This enabled us to create a FV vector toolbox for efficient, transient delivery (TraFo) of CRISPR/Cas9 components into different target tissues. Co-delivery of Cas9 mRNA by TraFo-Cas9 vectors in combination with retroviral, integration-deficient single guide RNA (sgRNA) expression enhanced efficacy and specificity of gene-inactivation compared with CRISPR/Cas9 lentiviral vector systems. Furthermore, separate TraFo-Cas9 delivery allowed the optional inclusion of a repair matrix for efficient gene correction or tagging as well as the addition of fluorescent negative selection markers for easy identification of off-target editing or incorrect repair events. Thus, the TraFo CRISPR toolbox represents an interesting alternative technology for gene inactivation and gene editing.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA