Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 234
Filtrar
1.
Front Immunol ; 12: 791206, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34804076

RESUMEN

Osteosarcoma, Ewing sarcoma (EWS), and rhabdomyosarcoma (RMS) are the most common pediatric sarcomas. Conventional therapy for these sarcomas comprises neoadjuvant and adjuvant chemotherapy, surgical resection of the primary tumor and/or radiation therapy. Patients with metastatic, relapsed, or refractory tumors have a dismal prognosis due to resistance to these conventional therapies. Therefore, innovative therapeutic interventions, such as immunotherapy, are urgently needed. Recently, cancer research has focused attention on natural killer (NK) cells due their innate ability to recognize and kill tumor cells. Osteosarcoma, EWS and RMS, are known to be sensitive to NK cell cytotoxicity in vitro. In the clinical setting however, NK cell cytotoxicity against sarcoma cells has been mainly studied in the context of allogeneic stem cell transplantation, where a rapid immune reconstitution of NK cells plays a key role in the control of the disease, known as graft-versus-tumor effect. In this review, we discuss the evidence for the current and future strategies to enhance the NK cell-versus-pediatric sarcoma effect, with a clinical focus. The different approaches encompass enhancing antibody-dependent NK cell cytotoxicity, counteracting the NK cell mechanisms of self-tolerance, and developing adoptive NK cell therapy including chimeric antigen receptor-expressing NK cells.


Asunto(s)
Neoplasias Óseas/inmunología , Inmunoterapia Adoptiva/métodos , Inmunoterapia/tendencias , Células Asesinas Naturales/inmunología , Osteosarcoma/inmunología , Rabdomiosarcoma/inmunología , Sarcoma de Ewing/inmunología , Animales , Anticuerpos/metabolismo , Neoplasias Óseas/terapia , Niño , Citotoxicidad Inmunológica , Efecto Injerto vs Tumor , Humanos , Activación de Linfocitos , Osteosarcoma/terapia , Rabdomiosarcoma/terapia , Sarcoma de Ewing/terapia
2.
J Orthop Res ; 39(3): 536-542, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33095470

RESUMEN

Immunotherapy of musculoskeletal tumors remains clinically challenging and requires the development of gene-engineered/adoptive exogenous immune cells or the identification of new molecular target(s) that can be therapeutically exploited to improve patient outcome. Recently, endogenous B-cell infiltration into tumor microenvironments appears to be an essential promising prognostic factor controlling tumor progression in musculoskeletal malignancy. Here, we explored the level of T-cell infiltration by analyzing expression profiles of CD3E, CD4, and CD8A in 1366 patients and 23 histological types. The data revealed that CD3E and CD8A expressions were predominantly inhibited in bone tumors when compared with normal bone. CD4 expression was upregulated in limited types of tumors, including chondrosarcoma and giant cell tumor of bone, whereas other tumors demonstrated relatively lower expressions. Similarly, regarding soft tissue sarcoma, the expression of T-cell-related molecules was largely inhibited. Only in patients with rhabdomyosarcoma, CD3E and CD8A expressions were significantly upregulated, showing the nature of immune-active tumor. To visualize the immunological microenvironment of rhabdomyosarcoma, we have developed a novel software aimed at analyzing numerous cell-to-cell and ligand-to-receptor interactions, that is, Environmentome. It has led to the identification of molecular interactions between CD8+ T cell and rhabdomyosarcoma via Galectin3-LAG3 binding, which is a novel immune checkpoint recently identified. In conclusion, musculoskeletal tumors may be defined as immune-quiescent tumors, whereby targeting Galectin-3 and/or immune-infiltrative agents could be crucial in these immunologically noninflamed musculoskeletal tumors, accelerating immunotherapeutic response.


Asunto(s)
Neoplasias de Tejido Conjuntivo/inmunología , Linfocitos T/fisiología , Linfocitos T CD8-positivos/fisiología , Humanos , Rabdomiosarcoma/inmunología
3.
Front Immunol ; 11: 581468, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33193388

RESUMEN

High-risk rhabdomyosarcoma (RMS) occurring in childhood to young adulthood is associated with a poor prognosis; especially children above the age of 10 with advanced stage alveolar RMS still succumb to the disease within a median of 2 years. The advent of chimeric antigen receptor (CAR)-engineered T cells marked significant progress in the treatment of refractory B cell malignancies, but experience for solid tumors has proven challenging. We speculate that this is at least in part due to the poor quality of the patient's own T cells and therefore propose using CAR-modified cytokine-induced killer (CIK) cells as effector cells. CIK cells are a heterogeneous population of polyclonal T cells that acquire phenotypic and cytotoxic properties of natural killer (NK) cells through the cultivation process, becoming so-called T-NK cells. CIK cells can be genetically modified to express CARs. They are minimally alloreactive and can therefore be acquired from haploidentical first-degree relatives. Here, we explored the potential of ERBB2-CAR-modified random-donor CIK cells as a treatment for RMS in xenotolerant mice bearing disseminated high-risk RMS tumors. In otherwise untreated mice, RMS tumors engrafted 13-35 days after intravenous tumor cell injection, as shown by in vivo bioluminescence imaging, immunohistochemistry, and polymerase chain reaction for human gDNA, and mice died shortly thereafter (median/range: 62/56-66 days, n = 5). Wild-type (WT) CIK cells given at an early stage delayed and eliminated RMS engraftment in 4 of 6 (67%) mice, while ERBB2-CAR CIK cells inhibited initial tumor load in 8 of 8 (100%) mice. WT CIK cells were detectable but not as active as CAR CIK cells at distant tumor sites. CIK cell therapies during advanced RMS delayed but did not inhibit tumor progression compared to untreated controls. ERBB2-CAR CIK cell therapy also supported innate immunity as evidenced by selective accumulation of NK and T-NK cell subpopulations in disseminated RMS tumors, which was not observed for WT CIK cells. Our data underscore the power of heterogenous immune cell populations (T, NK, and T-NK cells) to control solid tumors, which can be further enhanced with CARs, suggesting ERBB2-CAR CIK cells as a potential treatment for high-risk RMS.


Asunto(s)
Células Asesinas Inducidas por Citocinas/inmunología , Inmunidad Innata/inmunología , Células Asesinas Naturales/inmunología , Receptor ErbB-2/inmunología , Receptores Quiméricos de Antígenos/inmunología , Rabdomiosarcoma/inmunología , Adolescente , Animales , Línea Celular Tumoral , Humanos , Inmunoterapia Adoptiva/métodos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células T Asesinas Naturales/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Nat Commun ; 11(1): 3549, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32669548

RESUMEN

Refractory metastatic rhabdomyosarcoma is largely incurable. Here we analyze the response of a child with refractory bone marrow metastatic rhabdomyosarcoma to autologous HER2 CAR T cells. Three cycles of HER2 CAR T cells given after lymphodepleting chemotherapy induces remission which is consolidated with four more CAR T-cell infusions without lymphodepletion. Longitudinal immune-monitoring reveals remodeling of the T-cell receptor repertoire with immunodominant clones and serum autoantibodies reactive to oncogenic signaling pathway proteins. The disease relapses in the bone marrow at six months off-therapy. A second remission is achieved after one cycle of lymphodepletion and HER2 CAR T cells. Response consolidation with additional CAR T-cell infusions includes pembrolizumab to improve their efficacy. The patient described here is a participant in an ongoing phase I trial (NCT00902044; active, not recruiting), and is 20 months off T-cell infusions with no detectable disease at the time of this report.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Neoplasias de los Músculos/terapia , Recurrencia Local de Neoplasia/terapia , Receptor ErbB-2/inmunología , Rabdomiosarcoma/terapia , Linfocitos T/trasplante , Biopsia , Médula Ósea/patología , Niño , Ensayos Clínicos Fase I como Asunto , Humanos , Masculino , Neoplasias de los Músculos/inmunología , Neoplasias de los Músculos/patología , Recurrencia Local de Neoplasia/inmunología , Receptores Quiméricos de Antígenos/inmunología , Inducción de Remisión/métodos , Rabdomiosarcoma/inmunología , Rabdomiosarcoma/secundario , Linfocitos T/inmunología , Linfocitos T/metabolismo , Trasplante Autólogo/métodos , Resultado del Tratamiento
5.
Ann Clin Lab Sci ; 50(3): 412-416, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32581037

RESUMEN

Rhabdomyosarcoma (RMS) is a primitive embryonal mesenchymal neoplasm demonstrating skeletal muscle differentiation. Diagnosis of RMS remains difficult due to the diversity of clinical features, pathological forms, and lesion's locations. Immunohistochemistry and Fluorescence in Situ Hybridization are common methods used to aid RMS diagnosis. In this research we tested protein expression of Desmin (Clone MX046), MyoD1 (Clone MX049), MyoD1 (Clone 5.8A), MyoD1 (Clone EP212), Myogenin (Clone F5D), and cytogenetic features in 21 RMS cases, with following results: positive rates of Desmin (Clone MX046), MyoD1 (Clone MX049), MyoD1 (Clone 5.8A), MyoD1 (Clone EP212) and Myogenin (Clone F5D) were 100.00%, 100.00%, 90.48%, 95.24% and 85.71%, respectively, with cytoplasmic stains of MyoD1 (Clone 5.8A) in 38.10% (8/21) cases and only nuclear stains of MyoD1 (Clone EP212), MyoD1 (Clone MX049) in all positive cases. FOXO1 gene was detected apart in 9 alveolar RMS samples, where MyoD1 (Clone MX049), MyoD1 (Clone 5.8A) and MyoD1 (Clone EP212) were 100% positive but MyoD1 (Clone 5.8A) only 44.44% (4/9). Thus we believe MyoD1 (Clone MX049) performs more sensitive and specific than MyoD1 (Clone 5.8A) and MyoD1 (Clone EP212).


Asunto(s)
Anticuerpos Monoclonales/inmunología , Rabdomiosarcoma/diagnóstico , Rabdomiosarcoma/inmunología , Adolescente , Adulto , Anticuerpos Monoclonales/metabolismo , Biomarcadores de Tumor , China , Desmina/genética , Desmina/inmunología , Diagnóstico Diferencial , Femenino , Fluorescencia , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Masculino , Persona de Mediana Edad , Proteína MioD/inmunología , Proteína MioD/metabolismo , Miogenina/genética , Miogenina/inmunología , Rabdomiosarcoma/metabolismo
6.
Clin Cancer Res ; 26(15): 4018-4030, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32332015

RESUMEN

PURPOSE: Clinical trials with immune checkpoint inhibition in sarcomas have demonstrated minimal response. Here, we interrogated the tumor microenvironment (TME) of two contrasting soft-tissue sarcomas (STS), rhabdomyosarcomas and undifferentiated pleomorphic sarcomas (UPS), with differing genetic underpinnings and responses to immune checkpoint inhibition to understand the mechanisms that lead to response. EXPERIMENTAL DESIGN: Utilizing fresh and formalin-fixed, paraffin-embedded tissue from patients diagnosed with UPS and rhabdomyosarcomas, we dissected the TME by using IHC, flow cytometry, and comparative transcriptomic studies. RESULTS: Our results demonstrated both STS subtypes to be dominated by tumor-associated macrophages and infiltrated with immune cells that localized near the tumor vasculature. Both subtypes had similar T-cell densities, however, their in situ distribution diverged. UPS specimens demonstrated diffuse intratumoral infiltration of T cells, while rhabdomyosarcomas samples revealed intratumoral T cells that clustered with B cells near perivascular beds, forming tertiary lymphoid structures (TLS). T cells in UPS specimens were comprised of abundant CD8+ T cells exhibiting high PD-1 expression, which might represent the tumor reactive repertoire. In rhabdomyosarcomas, T cells were limited to TLS, but expressed immune checkpoints and immunomodulatory molecules which, if appropriately targeted, could help unleash T cells into the rest of the tumor tissue. CONCLUSIONS: Our work in STS revealed an immunosuppressive TME dominated by myeloid cells, which may be overcome with activation of T cells that traffic into the tumor. In rhabdomyosarcomas, targeting T cells found within TLS may be key to achieve antitumor response.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/farmacología , Neoplasias Complejas y Mixtas/inmunología , Rabdomiosarcoma/inmunología , Estructuras Linfoides Terciarias/inmunología , Macrófagos Asociados a Tumores/inmunología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Linfocitos B/inmunología , Linfocitos T CD8-positivos/inmunología , Niño , Preescolar , Resistencia a Antineoplásicos/inmunología , Femenino , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Persona de Mediana Edad , Neoplasias Complejas y Mixtas/tratamiento farmacológico , Neoplasias Complejas y Mixtas/genética , Neoplasias Complejas y Mixtas/patología , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/genética , Rabdomiosarcoma/patología , Estructuras Linfoides Terciarias/patología , Escape del Tumor , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Adulto Joven
7.
J Med Genet ; 57(2): 104-108, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-30352869

RESUMEN

BACKGROUND: Rhabdomyosarcoma (RMS) is rare cancer affecting children and adults. Pleomorphic RMS histology is almost exclusive to adult patients and often resistant to chemotherapy. OBJECTIVE: We report the case of a 19-year-old patient who presented with a metastatic chemoresistant pleomorphic RMS. METHODS: Considering the poor prognosis and the few systemic therapeutic options, we decided to carry out a whole exome sequencing (WES) of the tumour and germline DNA. RESULTS: WES identified a germline variation (c.1863_1864insT) in the MLH1 gene corresponding to a pathogenic mutation: (p. Leu622Serfs*10), whereas the family history did not fit with classical criteria for Lynch syndrome. Loss-of-heterozygosity at MLH1 locus was found in the tumour. Immunohistochemistry showed loss of MLH1 and PMS2 nuclear expression in the tumour cells. In view of the mismatch repair defects and a high programmed cell death ligand 1 (PD-L1) expression (60% of tumour cells expressed PD-L1), we administrated an anti-PD-1 antibody to the patient. He achieved a rapid complete response of the lung metastases, which appears sustained after a 1-year follow-up. CONCLUSION: This observation of an RMS revealing an unexpected Lynch syndrome underlines the overlap between tumorous and germline molecular genetics and emphasises the major impact of cancer genomic medicine in clinical practice for guiding treatment decision.


Asunto(s)
Antígeno B7-H1/inmunología , Neoplasias Pulmonares/terapia , Endonucleasa PMS2 de Reparación del Emparejamiento Incorrecto/genética , Homólogo 1 de la Proteína MutL/genética , Rabdomiosarcoma/terapia , Adulto , Anticuerpos Antiidiotipos , Antígeno B7-H1/antagonistas & inhibidores , Biomarcadores de Tumor/genética , Niño , Resistencia a Antineoplásicos/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal/genética , Humanos , Pérdida de Heterocigocidad/genética , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Rabdomiosarcoma/genética , Rabdomiosarcoma/inmunología , Rabdomiosarcoma/patología , Secuenciación del Exoma , Adulto Joven
8.
Artículo en Inglés | MEDLINE | ID: mdl-31826987

RESUMEN

OBJECTIVE: To report the presence of a new neuronal surface antibody against the metabotropic glutamate receptor 2 antibody (mGluR2-Ab) in 2 patients with paraneoplastic cerebellar ataxia. METHODS: mGluR2-Abs were initially characterized by immunohistochemistry on the rat brain and confirmed by immunofluorescence on HEK293 cells transfected with mGluR2. Additional studies included analysis of potential cross-reactivity with other mGluRs, expression of mGluR2 in patients' tumors, and the effects of mGluR2-Abs on cultures of rat hippocampal neurons. RESULTS: Patient 1 was a 78-year-old woman with progressive cerebellar ataxia with an initial relapsing-remitting course who developed a small-cell tumor of unknown origin. Patient 2 was a 3-year-old girl who presented a steroid-responsive acute cerebellitis preceding the diagnosis of an alveolar rhabdomyosarcoma. Patients' serum and CSF showed a characteristic immunostaining of the hippocampus and cerebellum in rat brain sections and immunolabeled the cell surface of live rat hippocampal neurons. HEK293 cells transfected with mGluR1, 2, 3, and 5 confirmed that patients' antibodies only recognized mGluR2. mGluR2-Abs were not detected in 160 controls, 120 with paraneoplastic, autoimmune, or degenerative ataxias, and 40 with autoimmune encephalitis and antibodies against mGluR5 or unknown antigens. Expression of mGluR2 in tumors was confirmed by immunohistochemistry using a commercial mGluR2-Ab. Incubation of live rat hippocampal neurons with CSF of patient 2 did not modify the density of surface mGluR2 clusters. CONCLUSIONS: mGluR2-Abs are a novel biomarker of paraneoplastic cerebellar ataxia. The potential pathogenic effect of the antibodies is not mediated by downregulation or internalization of neuronal surface mGluR2.


Asunto(s)
Autoanticuerpos , Ataxia Cerebelosa , Neoplasias , Síndromes Paraneoplásicos del Sistema Nervioso , Receptores de Glutamato Metabotrópico/inmunología , Anciano , Animales , Autoanticuerpos/inmunología , Autoanticuerpos/metabolismo , Biomarcadores de Tumor/inmunología , Biomarcadores de Tumor/metabolismo , Carcinoma Neuroendocrino/complicaciones , Carcinoma Neuroendocrino/inmunología , Carcinoma Neuroendocrino/metabolismo , Ataxia Cerebelosa/etiología , Ataxia Cerebelosa/inmunología , Ataxia Cerebelosa/metabolismo , Preescolar , Femenino , Células HEK293 , Humanos , Neoplasias Pulmonares/complicaciones , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/metabolismo , Neoplasias/complicaciones , Neoplasias/inmunología , Neoplasias/metabolismo , Síndromes Paraneoplásicos del Sistema Nervioso/etiología , Síndromes Paraneoplásicos del Sistema Nervioso/inmunología , Síndromes Paraneoplásicos del Sistema Nervioso/metabolismo , Ratas , Rabdomiosarcoma/complicaciones , Rabdomiosarcoma/inmunología , Rabdomiosarcoma/metabolismo
9.
Pathol Res Pract ; 215(12): 152700, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31704149

RESUMEN

BACKGROUND: Activation of immune checkpoints, e.g. PD-1/PD-L1 axis, in cancer microenvironment, enables evasion of host anti-cancer immune response and drives tumor progression. To date, there have been only a few studies analyzing PD-1/PD-L1 expression in pediatric malignancies. AIM: In the current study, we aimed to assess PD-L1 and PD-1 expression in pediatric rhabdomyosarcoma (RMS) and to investigate their clinicopathological associations. MATERIALS AND METHODS: The study enrolled 31 children with RMS. Tissue microarrays with representative tumor tissue samples were stained with anti-PD-1 NAT105 clone (Ventana, Roche) and two different antibodies against PD-L1: SP142 (Ventana, Roche) and 22C3 (DAKO). Adequate positive controls were applied. Their expression was assessed in tumor-associated immune cells (TAICs) and in the tumor cells separately. RESULTS: We did not detect any positive PD-L1 staining in analyzed tumors using SP142 antibody; however, in 11 cases (35.48%) its expression was revealed by means of 22C3 clone. The staining was restricted to TAICs in all cases, which no reaction in tumor cells. The 5-year relapse free survival (RFS) rate was significantly higher in PD-L1 positive cases (61.5% vs 25.0%, p = 0.024), but it most likely results from more frequent PD-L1 expression in low-stage RMS. PD-1 expression on TAICs was detected in 7 cases and did not influence the prognosis. CONCLUSIONS: We found that PD-L1 expression on TAICs, as detected with the use of 22C3 clone but not SP142 antibody, tends to be associated with low-stage RMS in children. PD-1 expression on TAICs in RMS is neither associated with distinct clinical course nor with clinicopathological features.


Asunto(s)
Antígeno B7-H1/análisis , Biomarcadores de Tumor/análisis , Inmunohistoquímica , Receptor de Muerte Celular Programada 1/análisis , Rabdomiosarcoma/inmunología , Adolescente , Edad de Inicio , Especificidad de Anticuerpos , Niño , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Estadificación de Neoplasias , Valor Predictivo de las Pruebas , Supervivencia sin Progresión , Rabdomiosarcoma/mortalidad , Rabdomiosarcoma/patología , Rabdomiosarcoma/terapia , Factores de Tiempo
10.
Front Immunol ; 10: 1814, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31428099

RESUMEN

Sarcoma is one of the most severe forms of pediatric cancer and current therapies -chemotherapy and surgery- fail to eradicate the disease in half of patients. Preclinical studies combining new therapeutic approaches can be useful to design better therapies. On one hand, it is known that CXCR4 expression is implicated in rhabdomyosarcoma progression, so we analyzed relapses and chemotherapy-resistant rhabdomyosarcoma tumors from pediatric patients and found that they had particularly high levels of CXCR4 expression. Moreover, in assays in vitro, anti-CXCR4 blocking antibody (MDX1338) efficiently reduced migration and invasion of alveolar rhabdomyosarcoma RH30 cells. On the other hand, activated and expanded natural killer (NKAE) cell therapy showed high cytotoxicity against sarcoma cells in vitro and completely inhibited RH30 tumor implantation in vivo. Only the combination of MDX1338 and NKAE treatments completely suppressed metastasis in mice. In this study, we propose a novel therapeutic approach based on anti-CXCR4 blocking antibody in combination with NKAE cell therapy to prevent rhabdomyosarcoma tumor implantation and lung metastasis. These results provide the first evidence for the efficacy of this combined immunotherapy for preventing sarcoma disease dissemination.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Células Asesinas Naturales/inmunología , Receptores CXCR4/inmunología , Sarcoma/inmunología , Sarcoma/terapia , Animales , Línea Celular Tumoral , Citotoxicidad Inmunológica/inmunología , Femenino , Humanos , Factores Inmunológicos/inmunología , Inmunoterapia , Inmunoterapia Adoptiva/métodos , Neoplasias Pulmonares/inmunología , Ratones , Ratones Endogámicos NOD , Rabdomiosarcoma/inmunología , Transducción de Señal/inmunología
11.
Cancer Res ; 79(13): 3455-3465, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31064845

RESUMEN

Compelling evidence points to immune cell infiltration as a critical component of successful immunotherapy. However, there are currently no clinically available, noninvasive methods capable of evaluating immune contexture prior to or during immunotherapy. In this study, we evaluate a T-cell-specific PET agent, [18F]F-AraG, as an imaging biomarker predictive of response to checkpoint inhibitor therapy. We determined the specificity of the tracer for activated T cells in vitro and in a virally induced model of rhabdomyosarcoma. Of all immune cells tested, activated human CD8+ effector cells showed the highest accumulation of [18F]F-AraG. Isolation of lymphocytes from the rhabdomyosarcoma tumors showed that more than 80% of the intratumoral signal came from accumulation of [18F]F-AraG in immune cells, primarily CD8+ and CD4+. Longitudinal monitoring of MC38 tumor-bearing mice undergoing anti-PD-1 treatment revealed differences in signal between PD-1 and isotype antibody-treated mice early into treatment. The differences in [18F]F-AraG signal were also apparent between responders and nonresponders to anti-PD-1 therapy. Importantly, we found that the signal in the tumor-draining lymph nodes provides key information about response to anti-PD-1 therapy. Overall, [18F]F-AraG has potential to serve as a much needed immunomonitoring clinical tool for timely evaluation of immunotherapy. SIGNIFICANCE: These findings reveal differences in T-cell activation between responders and nonresponders early into anti-PD-1 treatment, which may impact many facets of immuno-oncology, including patient selection, management, and development of novel combinatorial approaches.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Linfocitos T CD8-positivos/inmunología , Procesamiento de Imagen Asistido por Computador/métodos , Inmunoterapia , Activación de Linfocitos/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Rabdomiosarcoma/inmunología , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Femenino , Humanos , Activación de Linfocitos/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Ratones Endogámicos C57BL , Tomografía de Emisión de Positrones/métodos , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/metabolismo , Rabdomiosarcoma/patología , Células Tumorales Cultivadas
12.
Cell ; 177(7): 1903-1914.e14, 2019 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-31031007

RESUMEN

Xenograft cell transplantation into immunodeficient mice has become the gold standard for assessing pre-clinical efficacy of cancer drugs, yet direct visualization of single-cell phenotypes is difficult. Here, we report an optically-clear prkdc-/-, il2rga-/- zebrafish that lacks adaptive and natural killer immune cells, can engraft a wide array of human cancers at 37°C, and permits the dynamic visualization of single engrafted cells. For example, photoconversion cell-lineage tracing identified migratory and proliferative cell states in human rhabdomyosarcoma, a pediatric cancer of muscle. Additional experiments identified the preclinical efficacy of combination olaparib PARP inhibitor and temozolomide DNA-damaging agent as an effective therapy for rhabdomyosarcoma and visualized therapeutic responses using a four-color FUCCI cell-cycle fluorescent reporter. These experiments identified that combination treatment arrested rhabdomyosarcoma cells in the G2 cell cycle prior to induction of apoptosis. Finally, patient-derived xenografts could be engrafted into our model, opening new avenues for developing personalized therapeutic approaches in the future.


Asunto(s)
Animales Modificados Genéticamente/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias de los Músculos , Rabdomiosarcoma , Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente/genética , Animales Modificados Genéticamente/inmunología , Femenino , Xenoinjertos , Humanos , Células K562 , Masculino , Neoplasias de los Músculos/tratamiento farmacológico , Neoplasias de los Músculos/inmunología , Neoplasias de los Músculos/metabolismo , Neoplasias de los Músculos/patología , Trasplante de Neoplasias , Ftalazinas/farmacología , Piperazinas/farmacología , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/inmunología , Rabdomiosarcoma/metabolismo , Rabdomiosarcoma/patología , Temozolomida/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Pez Cebra/genética , Pez Cebra/inmunología
13.
Pediatr Hematol Oncol ; 36(1): 17-27, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30870043

RESUMEN

Long-term survival rates for pediatric patients with cancer have significantly improved, but novel approaches are desired for those with refractory/relapsed solid tumors. Recently, programed cell death-1/programed cell death-ligand-1 blockade has emerged as an effective option for many intractable cancers. However, not all patients show objective response to such therapy. On the other hand, several other checkpoint pathways, including Herpes virus entry mediator (HVEM)/B- and T-lymphocyte attenuator (BTLA), galectin-9 (GAL9)/T-cell immunoglobulin and mucin domain-3 (TIM3), and major histocompatibility complex class II (MHC-II)/lymphocyte activation gene-3 (LAG3), also regulate immune responses in the tumor microenvironment and may be alternative targets for novel immune therapies. In this study, we examined 65 common pediatric solid tumors and characterized the expression of Herpes virus entry mediator, GAL9, and MHC-II on tumor cells and their corresponding receptors B- and T-lymphocyte attenuator, TIM3, and LAG3, respectively, on tumor-infiltrating lymphocytes (TILs) with immunohistochemistry. Whereas the expression of GAL9 and MHC-II was limited, 73% of rhabdomyosarcomas and 100% of osteosarcomas expressed moderate to high levels of Herpes virus entry mediator on the tumor. TILs were detected in all tumor samples except one osteosarcoma. Interestingly, 45% of rhabdomyosarcomas, and 45% of osteosarcomas expressed moderate to high levels of both Herpes virus entry mediator on the tumor cells and B- and T-lymphocyte attenuator on the TILs. Results showed that a subset of pediatric solid tumors expressed tumor-associated checkpoint molecules, and TILs expressed corresponding receptors for these checkpoint molecules. Thus, immunogenic environments may be created, and checkpoint blockade may induce favorable immune responses.


Asunto(s)
Linfocitos B/inmunología , Neoplasias Óseas/inmunología , Proteínas de Neoplasias/inmunología , Osteosarcoma/inmunología , Rabdomiosarcoma/inmunología , Linfocitos T/inmunología , Adolescente , Linfocitos B/patología , Neoplasias Óseas/patología , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Osteosarcoma/patología , Rabdomiosarcoma/patología , Linfocitos T/patología
14.
BMC Cancer ; 19(1): 126, 2019 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-30732578

RESUMEN

BACKGROUND: Insulin-like Growth Factor Receptor-1 (IGF1R) system sustains the genesis of rhabdomyosarcoma through IGF2 autocrine overexpression. While several IGF1R-targeted strategies have been investigated to interphere with rhabdomyosarcoma growth, no attempt to neutralize IGF2 has been reported. We therefore studied the possibility to hamper rhabdomyosarcoma growth with passive and active immune approaches targeting IGF2. METHODS: A murine model developing IGF2-overexpressing pelvic rhabdomyosarcoma, along with IGF2-independent salivary carcinoma, was used to investigate the efficacy and specificity of passive anti-IGFs antibody treatment. Active vaccinations with electroporated DNA plasmids encoding murine or human IGF2 were performed to elicit autochthonous anti-IGF2 antibodies. Vaccinated mice received the intravenous injection of rhabdomyosarcoma cells to study the effects of anti-IGF2 antibodies against developing metastases. RESULTS: Passive administration of antibodies neutralizing IGFs delayed the onset of IGF2-overexpressing rhabdomyosarcoma but not of IGF2-independent salivary carcinoma. A DNA vaccine against murine IGF2 did not elicit antibodies, even when combined with Treg-depletion, while a DNA vaccine encoding the human IGF2 gene elicited antibodies crossreacting with murine IGF2. Mice with anti-IGF2 antibodies were partially protected against the metastatic growth of IGF2-addicted rhabdomyosarcoma cells. CONCLUSIONS: Immune targeting of autocrine IGF2 inhibited rhabdomyosarcoma genesis and metastatic growth.


Asunto(s)
Comunicación Autocrina , Inmunomodulación , Factor II del Crecimiento Similar a la Insulina/metabolismo , Rabdomiosarcoma/inmunología , Rabdomiosarcoma/metabolismo , Animales , Animales Modificados Genéticamente , Anticuerpos Monoclonales/farmacología , Antineoplásicos Inmunológicos/farmacología , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Humanos , Factor II del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Factor II del Crecimiento Similar a la Insulina/genética , Masculino , Ratones , Ratas , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/patología , Resultado del Tratamiento
15.
Oncogene ; 38(20): 3843-3854, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30670781

RESUMEN

Circular RNAs (circRNAs) represent a class of covalently closed RNAs, derived from non-canonical splicing events, which are expressed in all eukaryotes and often conserved among different species. We previously showed that the circRNA originating from the ZNF609 locus (circ-ZNF609) acts as a crucial regulator of human primary myoblast growth: indeed, the downregulation of the circRNA, and not of its linear counterpart, strongly reduced the proliferation rate of in vitro cultured myoblasts. To deepen our knowledge about circ-ZNF609 role in cell cycle regulation, we studied its expression and function in rhabdomyosarcoma (RMS), a pediatric skeletal muscle malignancy. We found that circ-ZNF609 is upregulated in biopsies from the two major RMS subtypes, embryonal (ERMS) and alveolar (ARMS). Moreover, we discovered that in an ERMS-derived cell line circ-ZNF609 knock-down induced a specific block at the G1-S transition, a strong decrease of p-Akt protein level and an alteration of the pRb/Rb ratio. Regarding p-Akt, we were able to show that circ-ZNF609 acts by counteracting p-Akt proteasome-dependent degradation, thus working as a new regulator of cell proliferation-related pathways. As opposed to ERMS-derived cells, the circRNA depletion had no cell cycle effects in ARMS-derived cells. Since in these cells the p53 gene resulted downregulated, with a concomitant upregulation of its cell cycle-related target genes, we suggest that this could account for the lack of circ-ZNF609 effect in ARMS.


Asunto(s)
ARN/genética , Rabdomiosarcoma/genética , Rabdomiosarcoma/patología , Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , ARN Circular , Rabdomiosarcoma/inmunología , Rabdomiosarcoma Alveolar/genética , Rabdomiosarcoma Alveolar/inmunología , Rabdomiosarcoma Alveolar/patología , Rabdomiosarcoma Embrionario/genética , Rabdomiosarcoma Embrionario/inmunología , Rabdomiosarcoma Embrionario/patología , Regulación hacia Arriba
16.
Cancer ; 123(19): 3807-3815, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28608950

RESUMEN

BACKGROUND: Programmed death 1 (PD-1) signaling in the tumor microenvironment dampens immune responses to cancer, and blocking this axis induces antitumor effects in several malignancies. Clinical studies of PD-1 blockade are only now being initiated in pediatric patients, and little is known regarding programmed death-ligand 1 (PD-L1) expression in common childhood cancers. The authors characterized PD-L1 expression and tumor-associated immune cells (TAICs) (lymphocytes and macrophages) in common pediatric cancers. METHODS: Whole slide sections and tissue microarrays were evaluated by immunohistochemistry for PD-L1 expression and for the presence of TAICs. TAICs were also screened for PD-L1 expression. RESULTS: Thirty-nine of 451 evaluable tumors (9%) expressed PD-L1 in at least 1% of tumor cells. The highest frequency histotypes comprised Burkitt lymphoma (80%; 8 of 10 tumors), glioblastoma multiforme (36%; 5 of 14 tumors), and neuroblastoma (14%; 17 of 118 tumors). PD-L1 staining was associated with inferior survival among patients with neuroblastoma (P = .004). Seventy-four percent of tumors contained lymphocytes and/or macrophages. Macrophages were significantly more likely to be identified in PD-L1-positive versus PD-L1-negative tumors (P < .001). CONCLUSIONS: A subset of diagnostic pediatric cancers exhibit PD-L1 expression, whereas a much larger fraction demonstrates infiltration with tumor-associated lymphocytes. PD-L1 expression may be a biomarker for poor outcome in neuroblastoma. Further preclinical and clinical investigation will define the predictive nature of PD-L1 expression in childhood cancers both at diagnosis and after exposure to chemoradiotherapy. Cancer 2017;123:3807-3815. © 2017 American Cancer Society.


Asunto(s)
Antígeno B7-H1/análisis , Linfocitos Infiltrantes de Tumor , Macrófagos , Proteínas de Neoplasias/análisis , Neoplasias/química , Neoplasias Óseas/química , Neoplasias Óseas/inmunología , Neoplasias Óseas/mortalidad , Neoplasias Óseas/patología , Linfoma de Burkitt/química , Linfoma de Burkitt/inmunología , Linfoma de Burkitt/patología , Niño , Glioblastoma/química , Glioblastoma/inmunología , Glioblastoma/patología , Humanos , Inmunohistoquímica , Neoplasias/inmunología , Neoplasias/mortalidad , Neoplasias/patología , Neuroblastoma/química , Neuroblastoma/inmunología , Neuroblastoma/mortalidad , Neuroblastoma/patología , Osteosarcoma/química , Osteosarcoma/inmunología , Osteosarcoma/patología , Rabdomiosarcoma/química , Rabdomiosarcoma/inmunología , Rabdomiosarcoma/patología , Sarcoma de Ewing/química , Sarcoma de Ewing/inmunología , Sarcoma de Ewing/patología , Análisis de Matrices Tisulares
17.
Sci Rep ; 7(1): 2396, 2017 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-28539588

RESUMEN

Oncolytic virotherapy is an effective immunotherapeutic approach for cancer treatment via a multistep process including direct tumor cell lysis, induction of cytotoxic or apoptosis-sensitizing cytokines and promotion of antitumor T cell responses. Solid tumors limit the effectiveness of immunotherapeutics in diverse ways such as secretion of immunosuppressive cytokines and expression of immune inhibitory ligands to inhibit antitumor T cell function. Blocking programmed cell death protein (PD)-1 signaling, which mediates T cell suppression via engagement of its inhibitory ligands, PD-L1 or PD-L2, is of particular interest due to recent successes in many types of cancer. In syngeneic murine rhabdomyosarcoma models, we found that M3-9-M (MHC I high) but not 76-9 (MHC I low) tumors respond to oncolytic herpes simplex virus-1 (oHSV-1) and PD-1 blockade combination therapy. In addition, the therapeutic outcomes in M3-9-M tumor models correlated with the increased incidence of CD4+ and CD8+ T cells but not with the CD4+CD25+Foxp3+ regulatory T cell populations in the tumor. Overall, our data suggest the combination of PD-1 blockade and oHSV-1 may be an effective treatment strategy for childhood soft tissue sarcoma.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Terapia Combinada/métodos , Viroterapia Oncolítica/métodos , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Rabdomiosarcoma/terapia , Simplexvirus/fisiología , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Citocinas/genética , Citocinas/inmunología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Femenino , Expresión Génica , Inyecciones Intralesiones , Inyecciones Intraperitoneales , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología , Rabdomiosarcoma/genética , Rabdomiosarcoma/inmunología , Rabdomiosarcoma/mortalidad , Análisis de Supervivencia , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología
18.
J Immunol ; 199(1): 82-90, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28550205

RESUMEN

The integrin α9ß1 is a key receptor involved in the development of autoimmune diseases. However, the detailed mechanism for the association of α9ß1 integrin with its ligands remains unclear. In this study, we introduce XCL1/lymphotactin, a member of the chemokine family, as a novel ligand for α9 integrin. Using α9 integrin-overexpressing NIH3T3 cells and endogenously α9 integrin-expressing human rhabdomyosarcoma cells, the interaction between XCL1 and α9 integrin was confirmed by pull-down assays. XCL1 enhanced α9 integrin-dependent cell migration of these cells, thus acting on α9 integrin as a chemoattractant. We also analyzed the in vivo function of XCL1 in the development of anti-type II collagen Ab-induced inflammatory arthritis (CAIA) in BALB/c mice and experimental autoimmune encephalomyelitis in C57BL/6 mice, because α9 integrin is involved in these autoimmune disease models. In CAIA, recombinant XCL1 aggravated the disease and this exacerbation was inhibited by an anti-α9 integrin Ab. An XCL1-neutralizing Ab produced in this study also ameliorated CAIA. Furthermore, the XCL1-neutralizing Ab abrogated the disease progression in experimental autoimmune encephalomyelitis. Therefore, to our knowledge this study provides the first in vitro and in vivo evidence that the interaction between XCL1 and α9 integrin has an important role for autoimmune diseases.


Asunto(s)
Quimiocinas C/inmunología , Quimiocinas C/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Cadenas alfa de Integrinas/metabolismo , Animales , Anticuerpos Neutralizantes/inmunología , Artritis Experimental/inmunología , Adhesión Celular , Movimiento Celular , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/fisiopatología , Encefalomielitis Autoinmune Experimental/terapia , Cadenas alfa de Integrinas/inmunología , Ligandos , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Rabdomiosarcoma/inmunología
19.
Oncotarget ; 8(2): 3495-3508, 2017 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-27966453

RESUMEN

Rhabdomyosarcoma (RMS), a neoplasm characterized by undifferentiated myoblasts, is the most common soft tissue tumour in children. Therapeutic resistance is common in RMS and is often caused by acquired defects in the cellular apoptotic program. Smac mimetic compounds (SMCs) are a novel class of inhibitor of apoptosis (IAP) antagonists that are currently under clinical development as cancer therapeutics. We previously reported that cIAP1 is overexpressed in human primary RMS tumours and in patient-derived RMS cell lines where it drives resistance to apoptosis. In this study, we investigated whether inflammatory cytokine production triggered by activators of innate immunity synergizes with LCL161 to induce bystander killing of RMS cells in vitro and in vivo. Indeed, we show that innate immune stimuli (oncolytic virus (VSVΔ51-GFP), interferon γ (IFNγ), and tumour necrosis factor-like weak inducer of apoptosis (TWEAK)) combine with SMCs in vitro to reduce cell viability in the Kym-1 RMS cancer cell line. Other human RMS cell lines (RH36, RH41, RD, RH18, RH28, and RH30) and the murine RMS cell line 76-9 are resistant to treatment with LCL161 alone or in combination with immune stimulants in in vitro cell viability assays. In contrast, we report that the combination of LCL161 and VSVΔ51-GFP reduces tumour volume and prolongs survival in a 76-9 syngeneic murine model. Our results support further exploration of the combined use of IAP antagonists and innate immune stimuli as a therapeutic approach for RMS cancers.


Asunto(s)
Imitación Molecular , Viroterapia Oncolítica , Virus Oncolíticos , Rabdomiosarcoma/inmunología , Rabdomiosarcoma/patología , Tiazoles/farmacología , Animales , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/inmunología , Terapia Combinada , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunidad Innata/efectos de los fármacos , Factor 1 Regulador del Interferón/genética , Factor 1 Regulador del Interferón/metabolismo , Ratones , Virus Oncolíticos/genética , Rabdomiosarcoma/genética , Rabdomiosarcoma/terapia , Carga Tumoral , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Clin Cancer Res ; 22(13): 3182-91, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-26823601

RESUMEN

PURPOSE: Patients with metastatic or relapsed pediatric sarcomas receive cytotoxic regimens that induce high remission rates associated with profound lymphocyte depletion, but ultimately few survive long term. We administered adjuvant immunotherapy to patients with metastatic and recurrent pediatric sarcomas in an effort to improve outcomes. EXPERIMENTAL DESIGN: Mononuclear cells were collected via apheresis, and tumor lysate was acquired via percutaneous biopsy at enrollment. Participants received standard antineoplastic therapy, followed by autologous lymphocytes, tumor lysate/keyhole limpet hemocyanin-pulsed dendritic cell vaccinations ± recombinant human IL7. Primary outcomes were toxicity and vaccine responses. Secondary outcomes were immune reconstitution, event-free survival, and overall survival (OS). RESULTS: Forty-three patients enrolled and 29 received immunotherapy. The regimen was well tolerated. Intent-to-treat analysis demonstrated 5-year OS of 51% with significant differences based upon histologic group (63% vs. 0% for Ewing/rhabdomyosarcoma vs. other sarcomas) and response to standard therapy (74% no residual disease vs. 0% residual disease). Five-year intent-to-treat OS of patients with newly diagnosed metastatic Ewing/rhabdomyosarcoma was 77%, higher than previously reported in this population and higher than observed in a similar group treated with an earlier adjuvant immunotherapy regimen (25% 5-year OS). T-cell responses to autologous tumor lysate were identified in 62% of immunotherapy recipients, and survival was higher in those patients (73% 5-year OS with vs. 37% without immune response, P = 0.017). Immune reconstitution, measured by CD4 count recovery, was significantly enhanced in subjects treated with recombinant human IL7. CONCLUSIONS: Adjuvant immunotherapy may improve survival in patients with metastatic pediatric sarcoma. Clin Cancer Res; 22(13); 3182-91. ©2016 AACR.


Asunto(s)
Células Dendríticas/trasplante , Factores Inmunológicos/uso terapéutico , Inmunoterapia Adoptiva/métodos , Interleucina-7/uso terapéutico , Rabdomiosarcoma/terapia , Sarcoma de Ewing/terapia , Linfocitos T/trasplante , Adolescente , Adulto , Quimioterapia Adyuvante/métodos , Niño , Preescolar , Terapia Combinada , Células Dendríticas/inmunología , Supervivencia sin Enfermedad , Femenino , Humanos , Leucaféresis , Masculino , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/prevención & control , Rabdomiosarcoma/inmunología , Rabdomiosarcoma/patología , Sarcoma de Ewing/inmunología , Sarcoma de Ewing/patología , Linfocitos T/inmunología , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...