Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Ther ; 29(12): 3410-3421, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34174441

RESUMEN

Natural killer (NK) cells mediate the cytolysis of transformed cells and are currently used as an adoptive cellular therapy to treat cancer. Infection with human cytomegalovirus has been shown to expand a subset of "adaptive" NK cells expressing the activation receptor NKG2C that have preferred functional attributes distinct from conventional NK cells. Because NKG2C delivers a strong activating signal to NK cells, we hypothesized that NKG2C could specifically trigger NK-cell-mediated antitumor responses. To elicit a tumor-directed response from NKG2C+ NK cells, we created an anti-NKG2C/IL-15/anti-CD33 killer engager called NKG2C-KE that directs NKG2C+ cells to target CD33+ cells and tumor-associated antigen expressed by acute myelogenous leukemia cells. The NKG2C-KE induced specific degranulation, interferon-γ production, and proliferation of NKG2C-expressing NK cells from patients who reactivated cytomegalovirus after allogeneic transplantation. The NKG2C-KE was also tested in a more homogeneous system using induced pluripotent stem cell (iPSC)-derived NK (iNK) cells that have been engineered to express NKG2C at high levels. The NKG2C-KE triggered iNK-cell-mediated cytotoxicity against CD33+ cells and primary AML blasts. The NKG2C-KE-specific interaction with adaptive NK and NKG2C+ iNK cells represents a new immunotherapeutic paradigm that uniquely engages highly active NK cells to induce cytotoxicity against AML through redirected targeting.


Asunto(s)
Células Madre Pluripotentes Inducidas , Leucemia Mieloide Aguda , Citomegalovirus , Humanos , Interleucina-15 , Células Asesinas Naturales , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/terapia
2.
Semin Immunol ; 31: 64-75, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28882429

RESUMEN

Natural killer (NK) cells have long been known to mediate anti-tumor responses without prior sensitization or recognition of specific tumor antigens. However, the tumor microenvironment can suppress NK cell function resulting in tumor escape and disease progression. Despite recent advances in cytokine therapy and NK cell adoptive transfer, tumor expression of ligands to NK - expressed checkpoint receptors can still suppress NK mediated tumor lysis. This review will explore many of the checkpoint receptors tumors utilize to manipulate the NK cell response as well as some of the current and upcoming pharmacological solutions to limit tumor suppression of NK cell function. Furthermore, we will discuss the potential to use these drugs in combinational therapies with novel antibody reagents such as bi- and tri-specific killer engagers (BiKEs and TriKEs) against tumor-specific antigens to enhance NK cell-mediated tumor rejection.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Vacunas contra el Cáncer/inmunología , Inmunoterapia Adoptiva/métodos , Células Asesinas Naturales/inmunología , Neoplasias/terapia , Animales , Terapia Combinada , Receptores Coestimuladores e Inhibidores de Linfocitos T/inmunología , Citotoxicidad Inmunológica , Humanos , Células Asesinas Naturales/trasplante , Neoplasias/inmunología
3.
Biol Blood Marrow Transplant ; 24(6): 1152-1162, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29505821

RESUMEN

Relapse is the most frequent cause of treatment failure after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Natural killer (NK) cells and γδ T cells reconstitute early after allo-HSCT, contribute to tumor immunosurveillance via major histocompatibility complex-independent mechanisms and do not induce graft-versus-host disease. Here we performed a quantitative and qualitative analysis of the NK and γδ T cell repertoire in healthy individuals, recipients of HLA-matched sibling or unrelated donor allo-HSCT (MSD/MUD-HSCT) and umbilical cord blood-HSCT (UCB-HSCT). NK cells are present at high frequencies in all allo-HSCT recipients. Immune reconstitution (IR) of vδ2+ cells depended on stem cell source. In MSD/MUD-HSCT recipients, vδ2+ comprise up to 8% of the total lymphocyte pool, whereas vδ2+ T cells are barely detectable in UCB-HSCT recipients. Vδ1+ IR was driven by CMV reactivation and was comparable between MSD/MUD-HSCT and UCB-HSCT. Strategies to augment NK cell mediated tumor responses, similar to IL-15 and antibodies, also induced vδ2+ T cell responses against a variety of different tumor targets. Vδ1+ γδ T cells were induced less by these same stimuli. We also identified elevated expression of the checkpoint inhibitory molecule TIGIT (T cell Ig and ITIM domain), which is also observed on tumor-infiltrating lymphocytes and epidermal γδ T cells. Collectively, these data show multiple strategies that can result in a synergized NK and γδ T cell antitumor response. In the light of recent developments of low-toxicity allo-HSCT platforms, these interventions may contribute to the prevention of early relapse.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/métodos , Reconstitución Inmune , Inmunoterapia/métodos , Linfocitos Intraepiteliales/citología , Células Asesinas Naturales/citología , Neoplasias/inmunología , Adolescente , Adulto , Estudios de Casos y Controles , Trasplante de Células Madre de Sangre del Cordón Umbilical , Humanos , Persona de Mediana Edad , Neoplasias/terapia , Prevención Secundaria/métodos , Hermanos , Trasplante Homólogo , Donante no Emparentado , Adulto Joven
4.
Mol Ther ; 24(7): 1312-22, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27157665

RESUMEN

Previously, we constructed a bispecific NK-cell-engager (BiKE) bearing single-chain variable fragments (scFv) against CD16 on NK cells and EpCAM on tumor cells. This BiKE facilitated antigen-specific antibody-dependent cell-mediated cytotoxicity (ADCC) but did not induce NK cell expansion. We incorporated a modified interleukin-15 cross-linker to create a trispecific construct (TriKE) in order to improve activation, proliferation, and survival of NK cells. Synthesis and assembly of hybrid genes encoding the TriKE was accomplished using DNA-shuffling and DNA-ligation techniques. The TriKE was tested for specificity, efficacy, proliferative capability, and cytokine profile using functional assays. The molecular modifications improved yield without compromising binding to EpCAM(+) HT-29 colorectal carcinoma cells. (51)Chromium-release and degranulation assays showed better killing rates with TriKE compared to BiKE. TriKE was more active in a variety of different carcinoma cell lines. TriKE showed the ability to stimulate expansion of CD56(+)CD3(-) NK cells. BiKE and TriKE showed enhanced but not supraphysiologic levels of cytokine secretion. 1615EpCAM TriKE drives enhanced ADCC while significantly improving proliferation, activation, and survival of NK cell effectors. The TriKE provides a selectively delivered self-sustaining signal at the NK/tumor cell synapse. Targeted cytokine stimulation, rather than systemic cytokine administration, may impact toxicity in patients rendering the TriKE a promising new off-the-shelf carcinoma therapy.


Asunto(s)
Anticuerpos Biespecíficos/genética , Citotoxicidad Celular Dependiente de Anticuerpos/genética , Carcinoma/inmunología , Interleucina-15/genética , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Activación de Linfocitos , Mutagénesis Insercional , Carcinoma/genética , Degranulación de la Célula/inmunología , Línea Celular Tumoral , Proliferación Celular , Citocinas/biosíntesis , Molécula de Adhesión Celular Epitelial/genética , Molécula de Adhesión Celular Epitelial/metabolismo , Expresión Génica , Orden Génico , Vectores Genéticos/genética , Humanos , Interferón gamma/genética , Interferón gamma/metabolismo , Unión Proteica , Proteínas Recombinantes de Fusión
5.
Blood ; 123(19): 3016-26, 2014 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-24652987

RESUMEN

Myelodysplastic syndromes (MDS) are stem cell disorders that can progress to acute myeloid leukemia. Although hematopoietic cell transplantation can be curative, additional therapies are needed for a disease that disproportionally afflicts the elderly. We tested the ability of a CD16xCD33 BiKE to induce natural killer (NK) cell function in 67 MDS patients. Compared with age-matched normal controls, CD7(+) lymphocytes, NK cells, and CD16 expression were markedly decreased in MDS patients. Despite this, reverse antibody-dependent cell-mediated cytotoxicity assays showed potent degranulation and cytokine production when resting MDS-NK cells were triggered with an agonistic CD16 monoclonal antibody. Blood and marrow MDS-NK cells treated with bispecific killer cell engager (BiKE) significantly enhanced degranulation and tumor necrosis factor-α and interferon-γ production against HL-60 and endogenous CD33(+) MDS targets. MDS patients had a significantly increased proportion of immunosuppressive CD33(+) myeloid-derived suppressor cells (MDSCs) that negatively correlated with MDS lymphocyte populations and CD16 loss on NK cells. Treatment with the CD16xCD33 BiKE successfully reversed MDSC immunosuppression of NK cells and induced MDSC target cell lysis. Lastly, the BiKE induced optimal MDS-NK cell function irrespective of disease stage. Our data suggest that the CD16xCD33 BiKE functions against both CD33(+) MDS and MDSC targets and may be therapeutically beneficial for MDS patients.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Células Asesinas Naturales/inmunología , Síndromes Mielodisplásicos/inmunología , Receptores de IgG/inmunología , Lectina 3 Similar a Ig de Unión al Ácido Siálico/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Biespecíficos/farmacología , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Células Cultivadas , Femenino , Citometría de Flujo , Células HL-60 , Humanos , Inmunosupresores/inmunología , Inmunosupresores/farmacología , Interferón gamma/inmunología , Interferón gamma/metabolismo , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/metabolismo , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/metabolismo , Síndromes Mielodisplásicos/prevención & control , Células Mieloides/efectos de los fármacos , Células Mieloides/inmunología , Células Mieloides/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Adulto Joven
6.
J Am Chem Soc ; 137(32): 10108-11, 2015 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-26230248

RESUMEN

Chimeric antigen receptors (CARs) have shown great promise for the immunological treatment of cancer. Nevertheless, the need to genetically engineer a patient's T-cells has presented significant production and safety challenges. To address these issues, we have demonstrated that chemically self-assembled nanorings (CSANs) displaying single chain antibodies can bind to both the CD3 ε subunit of the T-cell-receptor/CD3 complex and the CD22 antigen on malignant B cells such as B-leukemias or lymphomas. We demonstrate that the multivalent and bispecific format allows the antiCD3/antiCD22 CSANs to stably bind to T-cell surfaces for greater than 4 days, while being easily disassembled on the cell membrane by treatment with the nontoxic FDA approved drug, trimethoprim. In the presence of CD22+ Raji cells, T-cells modified with antiCD3/antiCD22 CSANs were shown to selectively up-regulate the production of interleukin-2 (IL-2) and interferon-γ (IFN-γ) and to initiate cytotoxicity. Taken together, our results demonstrate that antiCD3/antiCD22 bispecific CSANs offer a potential alternative to CARs, as prosthetic antigen receptors.


Asunto(s)
Complejo CD3/metabolismo , Ingeniería de Proteínas/métodos , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/farmacología , Anticuerpos Biespecíficos/metabolismo , Antígenos CD/inmunología , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/inmunología , Antígenos de Diferenciación de Linfocitos T/metabolismo , Complejo CD3/inmunología , Degranulación de la Célula , Citocinas/metabolismo , Dispersión Dinámica de Luz , Glicina/química , Humanos , Subunidad alfa del Receptor de Interleucina-2/inmunología , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Lectinas Tipo C/inmunología , Lectinas Tipo C/metabolismo , Activación de Linfocitos/efectos de los fármacos , Linfoma de Células B/inmunología , Linfoma de Células B/metabolismo , Linfoma de Células B/patología , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Receptores de Antígenos/química , Receptores de Antígenos/genética , Receptores de Antígenos/metabolismo , Lectina 2 Similar a Ig de Unión al Ácido Siálico/inmunología , Lectina 2 Similar a Ig de Unión al Ácido Siálico/metabolismo , Tetrahidrofolato Deshidrogenasa/metabolismo
7.
Int J Cancer ; 133(8): 1936-44, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-23553371

RESUMEN

Targeted toxins have the potential to overcome intrinsic or acquired resistance of cancer cells to conventional cytotoxic agents. Here, we hypothesized that EGFuPA-toxin, a bispecific ligand-targeted toxin (BLT) consisting of a deimmunized Pseudomonas exotoxin (PE) conjugated to epidermal growth factor and urokinase, would efficiently target and kill cells derived from canine hemangiosarcoma (HSA), a highly chemotherapy resistant tumor, as well as cultured hemangiospheres, used as a surrogate for cancer stem cells (CSC). EGFuPA-toxin showed cytotoxicity in four HSA cell lines (Emma, Frog, DD-1 and SB) at a concentration of ≤100 nM, and the cytotoxicity was dependent on specific ligand-receptor interactions. Monospecific targeted toxins also killed these chemoresistant cells; in this case, a "threshold" level of EGFR expression appeared to be required to make cells sensitive to the monospecific EGF-toxin, but not to the monospecific uPA-toxin. The IC50 of CSCs was higher by approximately two orders of magnitude as compared to non-CSCs, but these cells were still sensitive to EGFuPA-toxin at nanomolar (i.e., pharmacologically relevant) concentrations, and when targeted by EGFuPA-toxin, resulted in death of the entire cell population. Taken together, our results support the use of these toxins to treat chemoresistant tumors such as sarcomas, including those that conform to the CSC model. Our results also support the use of companion animals with cancer for further translational development of these cytotoxic molecules.


Asunto(s)
Proteínas Bacterianas/farmacología , Receptores ErbB/metabolismo , Hemangiosarcoma/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Animales , Anuros , Proteínas Bacterianas/toxicidad , Gatos , Línea Celular Tumoral , Citotoxinas/farmacología , Citotoxinas/toxicidad , Perros , Humanos , Inmunotoxinas/farmacología , Inmunotoxinas/toxicidad , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes de Fusión/toxicidad
8.
Gynecol Oncol ; 130(3): 579-87, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23721800

RESUMEN

OBJECTIVES: While most women with ovarian cancer will achieve complete remission after treatment, the majority will relapse within two years, highlighting the need for novel therapies. Cancer stem cells (CSC) have been identified in ovarian cancer and most other carcinomas as a small population of cells that can self-renew. CSC are more chemoresistant and radio-resistant than the bulk tumor cells; it is likely that CSC are responsible for relapse, the major problem in cancer treatment. CD133 has emerged as one of the most promising markers for CSC in ovarian cancer. The hypothesis driving this study is that despite their low numbers in ovarian cancer tumors, CSC can be eradicated using CD133 targeted therapy and tumor growth can be inhibited. METHODS: Ovarian cancer cell lines were evaluated using flow cytometry for expression of CD133. In vitro viability studies with an anti-CD133 targeted toxin were performed on one of the cell lines, NIH:OVCAR5. The drug was tested in vivo using a stably transfected luciferase-expressing NIH:OVCAR5 subline in nude mice, so that tumor growth could be monitored by digital imaging in real time. RESULTS: Ovarian cancer cell lines showed 5.6% to 16.0% CD133 expression. dCD133KDEL inhibited the in vitro growth of NIH:OVCAR5 cells. Despite low numbers of CD133-expressing cells in the tumor population, intraperitoneal drug therapy caused a selective decrease in tumor progression in intraperitoneal NIH:OVCAR5-luc tumors. CONCLUSIONS: Directly targeting CSC that are a major cause of drug resistant tumor relapse with an anti-CD133 targeted toxin shows promise for ovarian cancer therapy.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antígenos CD/metabolismo , Biomarcadores de Tumor/metabolismo , Citotoxinas/uso terapéutico , Glicoproteínas/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Neoplasias Ováricas/tratamiento farmacológico , Péptidos/metabolismo , Antígeno AC133 , Animales , Anticuerpos Monoclonales/farmacología , Antígenos CD/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citotoxinas/farmacología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Glicoproteínas/inmunología , Humanos , Ratones , Ratones Desnudos , Terapia Molecular Dirigida , Células Madre Neoplásicas/metabolismo , Neoplasias Ováricas/metabolismo , Péptidos/inmunología
9.
J Neurooncol ; 114(2): 155-64, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23695514

RESUMEN

Targeted toxins (TT) are molecules that bind cell surface antigens or receptors such as the transferrin or interleukin-13 receptor that are overexpressed in cancer. After internalization, the toxin component kills the cell. These recombinant proteins consist of an antibody or carrier ligand coupled to a modified plant or bacterial toxin such as diphtheria toxin (DT). These fusion proteins are very effective against brain cancer cells that are resistant to radiation therapy and chemotherapy. TT have shown an acceptable profile for toxicity and safety in animal studies and early clinical trials have demonstrated a therapeutic response. This review summarizes the characteristics of DT-based TT, the animal studies in malignant brain tumors and early clinical trial results. Obstacles to the successful treatment of brain tumors include poor penetration into tumor, the immune response to DT and cancer heterogeneity.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Toxina Diftérica/uso terapéutico , Inmunotoxinas/uso terapéutico , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/química , Toxinas Bacterianas/efectos adversos , Toxinas Bacterianas/química , Toxinas Bacterianas/uso terapéutico , Neoplasias Encefálicas/inmunología , Toxina Diftérica/efectos adversos , Toxina Diftérica/química , Humanos , Inmunotoxinas/administración & dosificación , Inmunotoxinas/efectos adversos , Ensayos Clínicos Controlados Aleatorios como Asunto , Proteínas Recombinantes de Fusión/efectos adversos , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/uso terapéutico , Transferrina/efectos adversos , Transferrina/química , Transferrina/uso terapéutico
10.
Mol Cancer Ther ; 22(3): 371-380, 2023 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-36548194

RESUMEN

Cancer stem-like cells (CSCs) are often the root cause of refractive relapse due to their inherent resistance to most therapies and ability to rapidly self-propagate. Recently, the antigen CD133 has been identified as a CSC marker on several cancer types and αCD133 therapies have shown selective targeting against CSCs with minimal off-target toxicity. Theoretically, by selectively eliminating CSCs, the sensitivity to bulk tumor-targeting therapies should be enhanced. Previously, our laboratory has developed bispecific chemically self-assembled nanorings (CSANs) that successfully induced T-cell eradication of EpCAM-positive (EpCAM+) tumors. We reasoned that targeting both CSCs [CD133-positive (CD133+)] and the bulk tumor (EpCAM+) simultaneously using our CSAN platform should produce a synergistic effect. We evaluated αCD133/αCD3 CSANs as both a single agent and in combination with αEpCAM/αCD3 CSANs to treat triple-negative breast cancer (TNBC) cells, which express a subpopulation of CD133+ cancer stem cells and EpCAM+ bulk tumor cells. Furthermore, an orthotopic breast cancer model validated the ability of αCD133 and αEpCAM targeting to combine synergistically in the elimination of TNBC MDA-MB-231 cells. Complete tumor eradication only occurred when EpCAM and CD133 were targeted simultaneously and lead to full remission in 80% of the test mice. Importantly, the depletion and enrichment of CD133 TNBCs highlighted the role of CD133+ cancer cells in regulating tumor growth and progression. Collectively, our results demonstrate that dual targeting with bispecific CSANs can be effective against heterogenous tumor cell populations and that elimination of primary and CD133+ CSCs may be necessary for eradication of at least a subset of TNBC.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Humanos , Animales , Ratones , Molécula de Adhesión Celular Epitelial , Neoplasias de la Mama Triple Negativas/patología , Línea Celular Tumoral , Linfocitos T , Células Madre Neoplásicas/patología , Antígeno AC133
11.
Front Immunol ; 14: 1060905, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36911670

RESUMEN

New treatments are required to enhance current therapies for lung cancer. Mesothelin is a surface protein overexpressed in non-small cell lung cancer (NSCLC) that shows promise as an immunotherapeutic target in phase I clinical trials. However, the immunosuppressive environment in NSCLC may limit efficacy of these therapies. We applied time-of-flight mass cytometry to examine the state of circulating mononuclear cells in fourteen patients undergoing treatment for unresectable lung cancer. Six patients had earlier stage NSCLC (I-IVA) and eight had highly advanced NSCLC (IVB). The advanced NSCLC patients relapsed with greater frequency than the earlier stage patients. Before treatment, patients with very advanced NSCLC had a greater proportion of CD14- myeloid cells than patients with earlier NSCLC. These patients also had fewer circulating natural killer (NK) cells bearing an Fc receptor, CD16, which is crucial to antibody-dependent cellular cytotoxicity. We designed a high affinity tri-specific killer engager (TriKE®) to enhance NK cytotoxicity against mesothelin+ targets in this environment. The TriKE consisted of CD16 and mesothelin binding elements linked together by IL-15. TriKE enhanced proliferation of lung cancer patient NK cells in vitro. Lung cancer lines are refractory to NK cell killing, but the TriKE enhanced cytotoxicity and cytokine production by patient NK cells when challenged with tumor. Importantly, TriKE triggered NK cell responses from patients at all stages of disease and treatment, suggesting TriKE can enhance current therapies. These pre-clinical studies suggest mesothelin-targeted TriKE has the potential to overcome the immunosuppressive environment of NSCLC to treat disease.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Neoplasias Pulmonares/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Mesotelina , Células Asesinas Naturales/metabolismo , Citotoxicidad Celular Dependiente de Anticuerpos , Inmunosupresores/metabolismo
12.
Blood ; 116(14): 2484-93, 2010 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-20570856

RESUMEN

Tumor-induced immune defects can weaken host immune response and permit tumor cell growth. In a systemic model of murine acute myeloid leukemia (AML), tumor progression resulted in increased regulatory T cells (Treg) and elevation of program death-1 (PD-1) expression on CD8(+) cytotoxic T cells (CTLs) at the tumor site. PD-1 knockout mice were more resistant to AML despite the presence of similar percentage of Tregs compared with wild type. In vitro, intact Treg suppression of CD8(+) T-cell responses was dependent on PD-1 expression by T cells and Tregs and PD-L1 expression by antigen-presenting cells. In vivo, the function of adoptively transferred AML-reactive CTLs was reduced by AML-associated Tregs. Anti-PD-L1 monoclonal antibody treatment increased the proliferation and function of CTLs at tumor sites, reduced AML tumor burden, and resulted in long-term survivors. Treg depletion followed by PD-1/PD-L1 blockade showed superior efficacy for eradication of established AML. These data demonstrated that interaction between PD-1 and PD-L1 can facilitate Treg-induced suppression of T-effector cells and dampen the antitumor immune response. PD-1/PD-L1 blockade coupled with Treg depletion represents an important new approach that can be readily translated into the clinic to improve the therapeutic efficacy of adoptive AML-reactive CTLs in advanced AML disease.


Asunto(s)
Antígenos de Superficie/inmunología , Proteínas Reguladoras de la Apoptosis/inmunología , Leucemia Mieloide Aguda/inmunología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Reguladores/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Antígenos de Superficie/genética , Proteínas Reguladoras de la Apoptosis/genética , Antígeno B7-1/inmunología , Antígeno B7-H1 , Antígenos CD8/inmunología , Regulación Leucémica de la Expresión Génica , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Depleción Linfocítica , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Péptidos/inmunología , Receptor de Muerte Celular Programada 1
13.
J Neurooncol ; 109(2): 229-38, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22696210

RESUMEN

The aim of this study is to investigate the anti-cancer effect of the bispecific diphtheria toxin (DT) based immunotoxin DTATEGF, which targets both the epidermal growth factor (EGF) receptor (EGFR) and the urokinase-type plasminogen activator (uPA) receptor (uPAR) in vitro and in vivo when delivered by convection-enhanced delivery (CED) via an osmotic minipump in a human metastatic non-small cell lung cancer (NSCLC) brain tumor mouse xenograft model. The effects of the bispecific immunotoxin DTATEGF, and monospecific DTAT, DTEGF and control DT at various concentrations were tested for their ability to inhibit the proliferation of human metastatic NSCLC PC9-BrM3 cells in vitro by MTT assay. A xenograft model of human metastatic NSCLC intracranial model was established in nude mice using the human NSCLC PC9-BrM3 cell line genetically marked with a firefly luciferase reporter gene. One microgram of DTATEGF in the treatment group or control DT in the control group was delivered intracranially by CED via an osmotic minipump. The bioluminescent imaging (BLI) was performed at day 7, 14, 1 month, 2 months, and 3 months. Kaplan-Meier survival curves for the two groups were generated. The brain tissue samples were stained by hematoxylin and eosin for histopathological assessment. In vitro, DTATEGF could selectively kill PC9-BrM3 cells and showed an IC(50) less than 0.001 nM, representing a more than 100- to 1000-fold increase in activity as compared to monospecific DTAT and DTEGF. In vivo, mice with tumors were treated intracranially with drug via CED where the results showed the treatment was successful in providing a survival benefit with the median survival of mice treated with DTATEGF being significantly prolonged relative to controls (87 vs. 63 days, P = 0.006). The results of these experiments indicate that DTATEGF kills the NSCLC PC9-BrM3 cell line in vitro, and when it is delivered via CED intracranially, it is highly efficacious against metastatic NSCLC brain tumors. DTATEGF is a safe and effective drug where further preclinical and clinical development is warranted for the management of metastatic brain tumors.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/secundario , Carcinoma de Pulmón de Células no Pequeñas/patología , Toxina Diftérica/administración & dosificación , Factor de Crecimiento Epidérmico/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Neoplasias Encefálicas/mortalidad , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Sistemas de Liberación de Medicamentos , Humanos , Estimación de Kaplan-Meier , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Proteínas Recombinantes de Fusión/metabolismo , Factores de Tiempo , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Blood ; 114(18): 3793-802, 2009 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-19724059

RESUMEN

Tumor-induced immune suppression can permit tumor cells to escape host immune resistance. To elucidate host factors contributing to the poor response of adoptively transferred tumor-reactive cytotoxic T lymphocytes (CTLs), we used a systemic model of murine acute myeloid leukemia (AML). AML progression resulted in a progressive regulatory T-cell (Treg) accumulation in disease sites. The adoptive transfer of in vitro-generated, potently lytic anti-AML-reactive CTLs failed to reduce disease burden or extend survival. Compared with non-AML-bearing hosts, transferred CTLs had reduced proliferation in AML sites of metastases. Treg depletion by a brief course of interleukin-2 diphtheria toxin (IL-2DT) transiently reduced AML disease burden but did not permit long-term survival. In contrast, IL-2DT prevented anti-AML CTL hypoproliferation, increased the number of transferred CTLs at AML disease sites, reduced AML tumor burden, and resulted in long-term survivors that sustained an anti-AML memory response. These data demonstrated that Tregs present at AML disease sites suppress adoptively transferred CTL proliferation, limiting their in vivo expansion, and Treg depletion before CTL transfer can result in therapeutic efficacy in settings of substantial pre-existing tumor burden in which antitumor reactive CTL infusion alone has proven ineffective.


Asunto(s)
Traslado Adoptivo , Leucemia Mieloide Aguda/terapia , Depleción Linfocítica , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/trasplante , Linfocitos T Reguladores , Animales , Proliferación Celular/efectos de los fármacos , Toxina Diftérica/farmacología , Modelos Animales de Enfermedad , Humanos , Memoria Inmunológica/efectos de los fármacos , Interleucina-2/farmacología , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/inmunología , Ratones , Ratones Noqueados , Proteínas Recombinantes de Fusión/farmacología , Escape del Tumor/efectos de los fármacos , Escape del Tumor/inmunología
16.
J Neurooncol ; 103(2): 255-66, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20830604

RESUMEN

A bispecific ligand-directed toxin (BLT), called EGFATFKDEL, consisting of human epidermal growth factor, a fragment of urokinase, and truncated pseudomonas exotoxin (PE38) was assembled in order to target human glioblastoma. Immunogenicity was reduced by mutating seven immunodominant B-cell epitopes on the PE38 molecule to create a new agent, EGFATFKDEL 7mut. In vitro, the drug selectively killed several human glioblastoma cell lines. EGFATFKDEL is our first BLT designed to simultaneously target EGFR on solid tumors and uPAR on the tumor neovasculature. In vitro assays revealed that the agent is effective against glioblastoma cell lines as well as human umbilical vein endothelial cells (HUVEC). Additionally, the bispecific drug displayed enhanced binding to overexpressed epidermal growth factor receptor and urokinase receptor when compared to similar monospecific drugs, EGFKDEL and ATFKDEL. In vivo, an aggressive human glioblastoma cell line was genetically marked with a firefly luciferase reporter gene and administered to the flanks of nude mice. Treatment with intratumoral injections of EGFATFKDEL 7mut eradicated small tumors in over half of the treated mice, which survived with tumor free status at least 100 days post tumor inoculation. ATFKDEL, which primarily targets the tumor neovasculature, prevented tumor growth but did not result in tumor-free mice in most cases. Specificity was shown by treating with an irrelevant BLT control which did not protect mice. Finally, immunization experiments in immunocompetent mice revealed significantly reduced anti-toxin production in EGFATFKDEL 7mut treated groups. Thus, EGFATFKDEL 7mut is an effective drug for glioblastoma therapy in this murine model and warrants further study.


Asunto(s)
ADP Ribosa Transferasas/farmacología , Antineoplásicos/farmacología , Toxinas Bacterianas/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Receptores ErbB , Exotoxinas/farmacología , Glioblastoma/tratamiento farmacológico , Activador de Plasminógeno de Tipo Uroquinasa , Factores de Virulencia/farmacología , Animales , Neoplasias Encefálicas/metabolismo , Células Endoteliales/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Citometría de Flujo , Glioblastoma/metabolismo , Humanos , Ligandos , Ratones , Ratones Desnudos , Fragmentos de Péptidos/síntesis química , Proteínas Recombinantes de Fusión/síntesis química , Proteínas Recombinantes de Fusión/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Exotoxina A de Pseudomonas aeruginosa
17.
Cancers (Basel) ; 13(16)2021 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-34439149

RESUMEN

Clinical studies validated antibodies directed against HER2, trastuzumab, and pertuzumab, as useful methodology to target breast cancer cases where HER2 is expressed. The hope was that HER2 targeting using these antibodies in ovarian cancer patients would prove useful as well, but clinical studies have shown lackluster results in this setting, indicating a need for a more comprehensive approach. Immunotherapy approaches stimulating the innate immune system show great promise, although enhancing natural killer (NK) function is not an established mainstream immunotherapy. This study focused on a new nanobody platform technology in which the bispecific antibody was altered to incorporate a cytokine. Herein we describe bioengineered CAM1615HER2 consisting of a camelid VHH antibody fragment recognizing CD16 and a single chain variable fragment (scFv) recognizing HER2 cross-linked by the human interleukin-15 (IL-15) cytokine. This tri-specific killer engager (TriKETM) showed in vitro prowess in its ability to kill ovarian cancer human cell lines. In addition, we demonstrated its efficacy in inducing potent anti-cancer effects in an in vivo xenograft model of human ovarian cancer engrafting both cancer cells and human NK cells. While previous approaches with trastuzumab and pertuzumab faltered in ovarian cancer, the hope is incorporating targeting and cytokine priming within the same molecule will enhance efficacy in this setting.

18.
Leukemia ; 35(6): 1586-1596, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33097838

RESUMEN

The low 5-year survival rate for patients with acute myeloid leukemia (AML), primarily caused due to disease relapse, emphasizes the need for better therapeutic strategies. Disease relapse is facilitated by leukemic stem cells (LSCs) that are resistant to standard chemotherapy and promote tumor growth. To target AML blasts and LSCs using natural killer (NK) cells, we have developed a trispecific killer engager (TriKETM) molecule containing a humanized anti-CD16 heavy chain camelid single-domain antibody (sdAb) that activates NK cells, an IL-15 molecule that drives NK-cell priming, expansion and survival, and a single-chain variable fragment (scFv) against human CLEC12A (CLEC12A TriKE). CLEC12A is a myeloid lineage antigen that is highly expressed by AML cells and LSCs, but not expressed by normal hematopoietic stem cells (HSCs), thus minimizing off-target toxicity. The CLEC12A TriKE induced robust NK-cell specific proliferation, enhanced NK-cell activation, and killing of both AML cell lines and primary patient-derived AML blasts in vitro while sparing healthy HSCs. Additionally, the CLEC12A TriKE was able to reduce tumor burden in preclinical mouse models. These findings highlight the clinical potential of the CLEC12A TriKE for the effective treatment of AML.


Asunto(s)
Inmunoterapia/métodos , Interleucina-15/metabolismo , Células Asesinas Naturales/inmunología , Lectinas Tipo C/inmunología , Leucemia Mieloide Aguda/terapia , Receptores de IgG/inmunología , Receptores Mitogénicos/inmunología , Anticuerpos de Dominio Único/farmacología , Animales , Apoptosis , Proliferación Celular , Femenino , Proteínas Ligadas a GPI/inmunología , Humanos , Lectinas Tipo C/antagonistas & inhibidores , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Pronóstico , Receptores Mitogénicos/antagonistas & inhibidores , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Blood Adv ; 5(4): 1069-1080, 2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33599743

RESUMEN

Expression of programmed cell death protein 1 (PD-1) on natural killer (NK) cells has been difficult to analyze on human NK cells. By testing commercial clones and novel anti-PD-1 reagents, we found expression of functional PD-1 on resting human NK cells in healthy individuals and reconstituting NK cells early after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Peripheral blood samples from healthy individuals and transplant recipients were stained for PD-1 expression using the commercial anti-PD-1 clone PD1.3.1.3, fluorescein isothiocyanate (FITC)-labeled pembrolizumab, or an FITC-labeled single-chain variable fragment (scFv) reagent made from pembrolizumab. These reagents identified low yet consistent basal PD-1 expression on resting NK cells, a finding verified by finding lower PD-1 transcripts in sorted NK cells compared with those in resting or activated T cells. An increase in PD-1 expression was identified on paired resting NK cells after allo-HSCT. Blockade of PD-1 on resting NK cells from healthy donors with pembrolizumab did not enhance NK function against programmed death-ligand 1 (PD-L1)-expressing tumor lines, but blocking with its scFv derivative resulted in a twofold increase in NK cell degranulation and up to a fourfold increase in cytokine production. In support of this mechanism, PD-L1 overexpression of K562 targets suppressed NK cell function. Interleukin-15 (IL-15) activity was potent and could not be further enhanced by PD-1 blockade. A similar increase in function was observed with scFv PD-1 blockade on resting blood NK cells after allo-HSCT. We identify the functional importance of the PD-1/PD-L1 axis on human NK cells in which blockade or activation to overcome inhibition will enhance NK cell-mediated antitumor control.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Neoplasias , Humanos , Células Asesinas Naturales , Activación de Linfocitos , Receptor de Muerte Celular Programada 1
20.
J Am Chem Soc ; 132(48): 17247-57, 2010 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-21077608

RESUMEN

A number of clever recombinant methodologies have been developed that recapitulate the valencies of IgG's (bivalent) and IgA's (tetravalent). Although higher synthetic valencies have been achieved by conjugation of either monoclonal antibodies or single-chain antibodies to nanoparticles and liposomes, a method for the preparation of recombinant antibodies with valencies similar to IgM's (decavalent) but considerably less than what is generally found after antibody particle conjugation has yet to be devised. Recently, we have developed a methodology for the design of bivalent Chemically Self-Assembled Antibody Nanorings (CSANs). We now report the crystal structure of the nanoring subunit composed of the E. coli DHFR dimer and a methotrexate dimerizer (MTX2-C9) containing a visible nine methylene linker and a protocol for the preparation of CSANs from this subunit with valencies similar to IgM's, ranging from 8-10 single chain antibodies (scFvs). The multivalent CSANs were reversibly assembled from a fusion protein dihydrofolate reductase (DHFR)-DHFR-antiCD3 scFv containing a single glycine linker between the two DHFR scaffolding proteins. We also demonstrate that, similar to the parental bivalent anti-CD3 monoclonal antibody (mAB), anti-CD3 CSANs selectively bind to CD3+ leukemia cells and undergo rapid internalization through a caveolin-independent pathway that requires cholesterol, actin polymerization, and protein tyrosine kinase activation. While treatment with the monoclonal antibody leads to T-cell activation and nearly complete loss (i.e., 90%) of the surface displayed T-cell receptor (TCR), only 25-30% of the TCR down regulate and no significant T-cell proliferation is observed after treatment of peripheral blood mononuclear cells (PBMCs) with anti-CD3 CSANs. Consistent with the proliferation findings, 15-25% less CD25 (IL-2 receptor) was found on the surface of PBMCs treated with either the polyvalent or bivalent anti-CD3 CSANs, respectively, than on PBMCs treated with the parental mAB. Comparative experiments with F(ab')2 derived from the mAB confirm that the activation of the T-cells by the mAB is dependent on the Fc domain, and thus interactions of the PBMC T-cells with accessory cells, such as macrophages. Taken together, our results demonstrate that anti-CD3 CSANs with valencies ranging from 2 to 8 could be employed for radionuclide, drug, or potentially oligonucleotide delivery to T-cells without, as has been observed for other antibody conjugated nanoparticles, the deleterious effects of activation observed for mAB. Further the CSAN construct may be adapted for the preparation of other multivalent scFvs.


Asunto(s)
Materiales Biomiméticos/química , Complejo CD3/inmunología , Diseño de Fármacos , Inmunoglobulina M/inmunología , Nanoestructuras/química , Anticuerpos de Cadena Única/química , Anticuerpos de Cadena Única/inmunología , Materiales Biomiméticos/síntesis química , Cristalografía por Rayos X , Humanos , Metotrexato/química , Modelos Moleculares , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Transporte de Proteínas , Anticuerpos de Cadena Única/metabolismo , Linfocitos T/citología , Linfocitos T/inmunología , Tetrahidrofolato Deshidrogenasa/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA