Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Cancer Immunol Immunother ; 72(5): 1153-1167, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36355079

RESUMEN

Multiple myeloma (MM) is an incurable hematological cancer, in which immune checkpoint inhibition (ICI) with monoclonal antibodies (mAbs) has failed due to uncontrollable immune responses in combination therapies and lack of efficacy in monotherapies. Although NK cell-specific checkpoint targets such as NKG2A and KIRs are currently being evaluated in clinical trials, the clinical impact of NK cells on the PD1 cascade is less well understood compared to T cells. Furthermore, while NK cells have effector activity within the TME, under continuous ligand exposure, NK cell dysfunctionality may occur due to interaction of PD1 and its ligand PD-L1. Due to above-mentioned factors, we designed novel NK cell specific PD1-based chimeric switch receptors (PD1-CSR) by employing signaling domains of DAP10, DAP12 and CD3ζ to revert NK cell inhibition and retarget ICI. PD1-CSR modified NK cells showed increased degranulation, cytokine secretion and cytotoxicity upon recognition of PD-L1+ target cells. Additionally, PD1-CSR+ NK cells infiltrated and killed tumor spheroids. While primary NK cells (pNK), expressing native PD1, showed decreased degranulation and cytokine production against PD-L1+ target cells by twofold, PD1-CSR+ pNK cells demonstrated increased activity upon PD-L1+ target cell recognition and enhanced antibody-dependent cellular cytotoxicity. PD1-CSR+ pNK cells from patients with MM increased degranulation and cytokine expression against autologous CD138+PD-L1+ malignant plasma cells. Taken together, the present results demonstrate that PD1-CSR+ NK cells enhance and sustain potent anti-tumor activity in a PD-L1+ microenvironment and thus represent a promising strategy to advance adoptive NK cell-based immunotherapies toward PD-L1+ cancers.


Asunto(s)
Antígeno B7-H1 , Mieloma Múltiple , Humanos , Antígeno B7-H1/metabolismo , Ligandos , Células Asesinas Naturales , Citocinas/metabolismo , Receptores de Células Asesinas Naturales/metabolismo , Inmunoterapia/métodos , Microambiente Tumoral
2.
Cytotherapy ; 25(7): 763-772, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37055320

RESUMEN

BACKGROUND AIMS: Adoptive cell therapy with chimeric antigen receptor (CAR)-expressing natural killer (NK) cells is an emerging approach that holds promise in multiple myeloma (MM). However, the generation of CAR-NK cells targeting CD38 is met with obstacles due to the expression of CD38 on NK cells. Knock-out of CD38 is currently explored as a strategy, although the consequences of the lack of CD38 expression with regards to engraftment and activity in the bone marrow microenvironment are not fully elucidated. Here, we present an alternative approach by harnessing the CD38dim phenotype occurring during long-term cytokine stimulation of primary NK cells. METHODS: Primary NK cells were expanded from peripheral blood mononuclear cells by long-term IL-2 stimulation. During expansion, the CD38 expression was monitored in order to identify a time point when introduction of a novel affinity-optimized αCD38-CAR confered optimal viability, i.e. prevented fratricide. CD38dim NK cells were trasduced with retroviral vectors encoding for the CAR trasngene and their functionality was assessed in in vitro activation and cytotoxicity assays. RESULTS: We verified the functionality of the αCD38-CAR-NK cells against CD38+ cell lines and primary MM cells. Importantly, we demonstrated that αCD38-CAR-NK cells derived from patients with MM have increased activity against autologous MM samples ex vivo. CONCLUSIONS: Overall, our results highlight that incorporation of a functional αCD38-CAR construct into a suitable NK-cell expansion and activation protocol results in a potent and feasible immunotherapeutic strategy for the treatment of patients with MM.


Asunto(s)
Mieloma Múltiple , Receptores Quiméricos de Antígenos , Humanos , Receptores Quiméricos de Antígenos/metabolismo , Citocinas/metabolismo , Mieloma Múltiple/terapia , Leucocitos Mononucleares/metabolismo , Células Asesinas Naturales , Fenotipo , Inmunoterapia , Inmunoterapia Adoptiva/métodos , Línea Celular Tumoral , Microambiente Tumoral
3.
Eur J Haematol ; 106(4): 546-554, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33471414

RESUMEN

INTRODUCTION: Novel drugs and drug combinations have improved outcomes for multiple myeloma patients. However, subgroups of patients still have a poor progression-free survival (PFS) and overall survival (OS). In an attempt to identify how the novel drugs affect the outcome in standard-risk and high-risk patients, respectively, we have investigated 715 multiple myeloma (MM) patients who have undergone high dose treatment followed by autologous stem cell transplantation at our center during 1995 - 2020. Outcomes during three time periods, 1995-1999 (period I), 2000-2009 (period II), and 2010-2020 (period III), were compared separately for standard-risk and high-risk patients. Risk stratification was based on chromosome analysis for periods II and III. RESULTS: The whole cohort of patients showed significantly improved OS with time during the three periods being at a median of 5.8, 7.0, and 10.0 years, respectively. There is also a weak tendency for improved PFS, that is, a median of 2.4, 2.6, and 2.9 years, respectively, during the same periods. However, the separate analysis of standard-risk and high-risk patients showed that the overall improvement with time was due to improved standard-risk patients (median OS 8.4 years for the period I and not reached for period II and III). In contrast, no significant improvement was seen in high-risk patients. For patients with del17p, PFS was even worse during period III as compared to period II (median 1.6 vs 3.2 years respectively). CONCLUSION: Our results show that the dramatic improvement in outcome for MM patients during the last 20 years only applies for standard-risk patients, while high-risk MM patients still are doing poorly, indicating that the novel drugs developed during this time are preferentially effective in standard-risk patients. New treatment modalities like CAR-T cells, CAR-NK cells, and/or bispecific antibodies should be tried in clinical studies early in the course of the disease, especially in patients with high-risk cytogenetics.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Mieloma Múltiple/mortalidad , Mieloma Múltiple/terapia , Adulto , Anciano , Aberraciones Cromosómicas , Terapia Combinada , Análisis Citogenético , Femenino , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Hibridación Fluorescente in Situ , Masculino , Persona de Mediana Edad , Mieloma Múltiple/diagnóstico , Mieloma Múltiple/etiología , Pronóstico , Retratamiento , Análisis de Supervivencia , Trasplante Autólogo , Resultado del Tratamiento
4.
Eur J Haematol ; 105(6): 751-754, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32745304

RESUMEN

COVID-19 has emerged as a global pandemic. Cancer patients have been reported to be at higher risk for adverse outcome of COVID-19. Studies are ongoing to decipher the risk factors and risk groups among cancer patients as well as strategies to refine treatment approaches. Here, we report eight patients with multiple myeloma that underwent immunomodulatory therapies with daratumumab or lenalidomide-based combination treatments and one patient with smoldering multiple myeloma, all of which presented with symptomatic COVID-19. We report that patients that succumbed to COVID-19 presented with either progressive tumor disease under daratumumab treatment or were in remission under lenalidomide-dexamethasone treatment.


Asunto(s)
Anticuerpos Monoclonales/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Infecciones por Coronavirus/diagnóstico , Dexametasona/efectos adversos , Lenalidomida/efectos adversos , Mieloma Múltiple/diagnóstico , Neumonía Viral/diagnóstico , Talidomida/efectos adversos , Anticuerpos Monoclonales/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Betacoronavirus/patogenicidad , COVID-19 , Estudios de Cohortes , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/mortalidad , Dexametasona/administración & dosificación , Progresión de la Enfermedad , Femenino , Humanos , Inmunomodulación , Lenalidomida/administración & dosificación , Masculino , Persona de Mediana Edad , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/inmunología , Mieloma Múltiple/mortalidad , Pandemias , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/inmunología , Neumonía Viral/mortalidad , Pronóstico , Inducción de Remisión , SARS-CoV-2 , Índice de Severidad de la Enfermedad , Análisis de Supervivencia , Talidomida/administración & dosificación , Resultado del Tratamiento
5.
Proc Natl Acad Sci U S A ; 113(16): 4476-81, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27044098

RESUMEN

Inhibition of the vascular endothelial growth factor (VEGF) pathway has failed to improve overall survival of patients with glioblastoma (GBM). We previously showed that angiopoietin-2 (Ang-2) overexpression compromised the benefit from anti-VEGF therapy in a preclinical GBM model. Here we investigated whether dual Ang-2/VEGF inhibition could overcome resistance to anti-VEGF treatment. We treated mice bearing orthotopic syngeneic (Gl261) GBMs or human (MGG8) GBM xenografts with antibodies inhibiting VEGF (B20), or Ang-2/VEGF (CrossMab, A2V). We examined the effects of treatment on the tumor vasculature, immune cell populations, tumor growth, and survival in both the Gl261 and MGG8 tumor models. We found that in the Gl261 model, which displays a highly abnormal tumor vasculature, A2V decreased vessel density, delayed tumor growth, and prolonged survival compared with B20. In the MGG8 model, which displays a low degree of vessel abnormality, A2V induced no significant changes in the tumor vasculature but still prolonged survival. In both the Gl261 and MGG8 models A2V reprogrammed protumor M2 macrophages toward the antitumor M1 phenotype. Our findings indicate that A2V may prolong survival in mice with GBM by reprogramming the tumor immune microenvironment and delaying tumor growth.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Anticuerpos Antineoplásicos/farmacología , Antineoplásicos/farmacología , Glioblastoma/tratamiento farmacológico , Macrófagos/metabolismo , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias Experimentales/tratamiento farmacológico , Ribonucleasa Pancreática/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Proteínas de Transporte Vesicular/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Macrófagos/patología , Ratones , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Ribonucleasa Pancreática/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Strahlenther Onkol ; 194(11): 975-984, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29855667

RESUMEN

PURPOSE: To ascertain the optimal radiation technique and radiation position for the neoadjuvant radiotherapy of patients with rectal cancer. MATERIALS AND METHODS: Treatment plans with similar dose objectives were generated for 20 selected patients. Dosimetric comparison was performed between prone and supine positions and between different radiation techniques. Dosimetric indices for the target volume and organs at risk (OAR) as well as normal tissue complication probability (NTCP) of late small bowel toxicity were analyzed. RESULTS: The helical tomotherapy (HT) in the prone position provided the optimal dose homogeneity in the target volume with the value of 0. Superior conformity values were obtained for Sliding Window (SW), Rapid Arc (RA) and HT compared to three-dimensional conformal radiotherapy (3D-CRT) techniques. All of the techniques showed dose reduction to OAR in the high-dose area in prone position versus supine position. Pairwise comparison revealed significantly higher small bowel protection by RA in the prone position in the high-dose area (V75, V45Gy). Similarly, superior bladder sparing was found for 3D-CRT in the prone position at higher doses (V50, V75). More healthy tissue in the radiation volume was involved by application of 3D-CRT with no relevant difference between positions. The mean values of NTCP for the small bowel did not show clinically meaningful variation between the techniques. CONCLUSION: All techniques provided superior sparing of OAR in the prone position. At higher radiation doses, treatment in prone position resulted in significant OAR protection, especially concerning small bowel sparing by RA and bladder sparing by 3D CRT.


Asunto(s)
Planificación de Atención al Paciente , Posición Prona , Planificación de la Radioterapia Asistida por Computador , Radioterapia Conformacional , Radioterapia de Intensidad Modulada , Neoplasias del Recto/radioterapia , Posición Supina , Femenino , Humanos , Intestino Delgado/efectos de la radiación , Masculino , Persona de Mediana Edad , Terapia Neoadyuvante , Órganos en Riesgo/efectos de la radiación , Traumatismos por Radiación/etiología , Traumatismos por Radiación/prevención & control , Dosificación Radioterapéutica , Neoplasias del Recto/patología , Factores de Riesgo , Vejiga Urinaria/efectos de la radiación
7.
Methods Cell Biol ; 178: 43-61, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37516528

RESUMEN

Understanding the anti-tumor activity of immune cells and testing cancer immunotherapies requires conditions that are as life-like as possible. The tumor microenvironment (TME) describes a complex sum of cellular and acellular actors that influence both immune cells and tumor cells as well as their interplay. Yet in development phases of new immunotherapies, the screening of drugs and adoptive cell products benefits from reproducible and controlled conditions. Two-dimensional (2D) cell cultures cannot simultaneously meet these two challenges therefore lacking considerably predictive power owing to their artificial nature. Various 3D tumor models have therefore been implemented to mimic the architecture and intrinsic heterogeneity of a microtumor. This protocol provides an easy-to-follow, time-efficient, material-limited method for live cell killing and infiltration of single tumor spheroids. It uses multicellular tumor spheroids grown scaffold-free and allows co-culture with immune cells. This protocol is optimized for natural killer (NK) cell functionality assays. However, it can be transferred to other immune cells, in particular cytotoxic T cells. This assay can be analysed using life cell imaging (here with the IncuCyte S3 system) and/or flow cytometry.


Asunto(s)
Microscopía , Neoplasias , Humanos , Citometría de Flujo , Neoplasias/terapia , Neoplasias/patología , Células Asesinas Naturales/patología , Técnicas de Cocultivo , Esferoides Celulares , Microambiente Tumoral
8.
Front Immunol ; 9: 2159, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30319622

RESUMEN

Chemokines govern leukocyte migration by attracting cells that express their cognate ligands. Many cancer types show altered chemokine secretion profiles, favoring the recruitment of pro-tumorigenic immune cells and preventing the accumulation of anti-tumorigenic effector cells. This can ultimately result in cancer immune evasion. The manipulation of chemokine and chemokine-receptor signaling can reshape the immunological phenotypes within the tumor microenvironment in order to increase the therapeutic efficacy of cancer immunotherapy. Here we discuss the three chemokine-chemokine receptor axes, CXCR1/2-CXCL1-3/5-8, CXCR3-CXCL9/10/11, and CXCR4-CXCL12 and their role on pro-tumorigenic immune cells and anti-tumorigenic effector cells in solid tumors. In particular, we summarize current strategies to target these axes and discuss their potential use in treatment approaches.


Asunto(s)
Quimiocinas CXC/metabolismo , Inmunidad Celular , Neoplasias/inmunología , Receptores CXCR/metabolismo , Microambiente Tumoral/inmunología , Carcinogénesis/inmunología , Quimiocinas CXC/inmunología , Humanos , Inmunoterapia/métodos , Linfocitos/inmunología , Linfocitos/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Células Supresoras de Origen Mieloide/inmunología , Invasividad Neoplásica/inmunología , Neoplasias/patología , Neoplasias/terapia , Receptores CXCR/inmunología , Escape del Tumor/inmunología
9.
Matrix Biol ; 67: 47-62, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29360499

RESUMEN

Hematopoietic stem and progenitor cell (HSPC) functions are regulated by a specialized microenvironment in the bone marrow - the hematopoietic stem cell niche - of which the extracellular matrix (ECM) is an integral component. We describe here the localization of ECM molecules, in particular the laminin α4, α3 and α5 containing isoforms in the bone marrow. Laminin 421 (composed of laminin α4, ß2, γ1 chains) is identified as a major component of the bone marrow ECM, occurring abundantly surrounding venous sinuses and in a specialized reticular fiber network of the intersinusoidal spaces of murine bone marrow (BM) in close association with HSPC. Bone marrow from Lama4-/- mice is significantly less efficient in reconstituting the hematopoietic system of irradiated wildtype (WT) recipients in competitive bone marrow transplantation assays and shows reduced colony formation in vitro. This is partially due to retention of Lin-c-kit+Sca-1+CD48- long-term and short-term hematopoietic stem cells (LT-HSC/ST-HSC) in the G0 phase of the cell cycle in Lama4-/- bone marrow and hence a more quiescent phenotype. In addition, the extravasation of WT BM cells into Lama4-/- bone marrow is impaired, influencing the recirculation of HSPC. Our data suggest that these effects are mediated by a compensatory expression of laminin α5 containing isoforms (laminin 521/522) in Lama4-/- bone marrow. Collectively, these intrinsic and extrinsic effects lead to reduced HSPC numbers in Lama4-/- bone marrow and reduced hematopoietic potential.


Asunto(s)
Médula Ósea/metabolismo , Células Madre Hematopoyéticas/citología , Laminina/metabolismo , Animales , Ciclo Celular , Movimiento Celular , Matriz Extracelular/metabolismo , Células Madre Hematopoyéticas/metabolismo , Laminina/genética , Ratones , Nicho de Células Madre
10.
Oncol Res Treat ; 40(5): 244-252, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28448985

RESUMEN

Total-skin electron beam therapy (TSEBT) is one of most effective treatments that has been used for cutaneous T-cell lymphoma. Low-dose TSEBT regimens (10-12 Gy) appear to be an effective alternative to conventional-dose TSEBT (30-36 Gy), yielding short-term remission of cutaneous manifestations with minimal toxicity. TSEBT can be administered to patients any time after a diagnosis of mycosis fungoides (MF). Patients requiring rapid relief from cutaneous lesions or symptoms may particularly benefit from TSEBT as an initial therapy. Radiotherapy (RT) dose, boost radiation delivery, maintenance treatment, and radiation tolerability may enhance remission rates and improve relapse-free survival following TSEBT. In addition, salvage local RT or TSEBT may be safely applied with high effectiveness. In this review, we focus on the use of TSEBT in patients with several forms of primary cutaneous T-cell lymphoma, and highlight the potential of low-dose TSEBT as part of a promising therapeutic approach.


Asunto(s)
Electrones/uso terapéutico , Linfoma Cutáneo de Células T/patología , Linfoma Cutáneo de Células T/terapia , Traumatismos por Radiación/prevención & control , Radioterapia de Alta Energía/métodos , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/terapia , Terapia Combinada/efectos adversos , Terapia Combinada/métodos , Relación Dosis-Respuesta en la Radiación , Electrones/efectos adversos , Medicina Basada en la Evidencia , Femenino , Humanos , Masculino , Traumatismos por Radiación/etiología , Dosificación Radioterapéutica , Radioterapia de Alta Energía/efectos adversos , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA