Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Cell ; 178(3): 585-599.e15, 2019 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-31303383

RESUMEN

New opportunities are needed to increase immune checkpoint blockade (ICB) benefit. Whereas the interferon (IFN)γ pathway harbors both ICB resistance factors and therapeutic opportunities, this has not been systematically investigated for IFNγ-independent signaling routes. A genome-wide CRISPR/Cas9 screen to sensitize IFNγ receptor-deficient tumor cells to CD8 T cell elimination uncovered several hits mapping to the tumor necrosis factor (TNF) pathway. Clinically, we show that TNF antitumor activity is only limited in tumors at baseline and in ICB non-responders, correlating with its low abundance. Taking advantage of the genetic screen, we demonstrate that ablation of the top hit, TRAF2, lowers the TNF cytotoxicity threshold in tumors by redirecting TNF signaling to favor RIPK1-dependent apoptosis. TRAF2 loss greatly enhanced the therapeutic potential of pharmacologic inhibition of its interaction partner cIAP, another screen hit, thereby cooperating with ICB. Our results suggest that selective reduction of the TNF cytotoxicity threshold increases the susceptibility of tumors to immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Inmunoterapia , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Apoptosis/efectos de los fármacos , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Humanos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Interferón gamma/metabolismo , Estimación de Kaplan-Meier , Masculino , Ratones , Ratones Endogámicos C57BL , Neoplasias/mortalidad , Neoplasias/terapia , ARN Guía de Kinetoplastida/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Transducción de Señal/efectos de los fármacos , Factor 2 Asociado a Receptor de TNF/deficiencia , Factor 2 Asociado a Receptor de TNF/genética , Factor de Necrosis Tumoral alfa/farmacología , Receptor de Interferón gamma
3.
Mol Ther ; 26(6): 1482-1493, 2018 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-29735366

RESUMEN

Adoptive cell therapy (ACT) is becoming a prominent alternative therapeutic treatment for cancer patients relapsing on traditional therapies. In parallel, antibodies targeting immune checkpoint molecules, such as cytotoxic-T-lymphocyte-associated antigen 4 (CTLA-4) and cell death protein 1 pathway (PD-1), are rapidly being approved for multiple cancer types, including as first line therapy for PD-L1-expressing non-small-cell lung cancer. The combination of ACT and checkpoint blockade could substantially boost the efficacy of ACT. In this study, we generated a novel self-delivering small interfering RNA (siRNA) (sdRNA) that knocked down PD-1 expression on healthy donor T cells as well as patient-derived tumor-infiltrating lymphocytes (TIL). We have developed an alternative chemical modification of RNA backbone for improved stability and increased efficacy. Our results show that T cells treated with sdRNA specific for PD-1 had increased interferon γ (IFN-γ) secreting capacity and that this modality of gene expression interference could be utilized in our rapid expansion protocol for production of TIL for therapy. TIL expanded in the presence of PD-1-specific sdRNA performed with increased functionality against autologous tumor as compared to control TIL. This method of introducing RNAi into T cells to modify the expression of proteins could easily be adopted into any ACT protocol and will lead to the exploration of new combination therapies.


Asunto(s)
Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/terapia , Melanoma/inmunología , Melanoma/terapia , Receptor de Muerte Celular Programada 1/metabolismo , Linfocitos T/metabolismo , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Citometría de Flujo , Células HeLa , Humanos , Inmunoterapia Adoptiva/métodos , Interferón gamma/genética , Interferón gamma/metabolismo , Neoplasias Pulmonares/inmunología , Melanoma/metabolismo , Receptor de Muerte Celular Programada 1/genética , Interferencia de ARN/fisiología
4.
J Immunol ; 196(2): 759-66, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26673145

RESUMEN

Treatment of cancer patients by adoptive T cell therapy has yielded promising results. In solid tumors, however, T cells encounter a hostile environment, in particular with increased inflammatory activity as a hallmark of the tumor milieu that goes along with abundant reactive oxygen species (ROS) that substantially impair antitumor activity. We present a strategy to render antitumor T cells more resilient toward ROS by coexpressing catalase along with a tumor specific chimeric Ag receptor (CAR) to increase their antioxidative capacity by metabolizing H2O2. In fact, T cells engineered with a bicistronic vector that concurrently expresses catalase, along with the CAR coexpressing catalase (CAR-CAT), performed superior over CAR T cells as they showed increased levels of intracellular catalase and had a reduced oxidative state with less ROS accumulation in both the basal state and upon activation while maintaining their antitumor activity despite high H2O2 levels. Moreover, CAR-CAT T cells exerted a substantial bystander protection of nontransfected immune effector cells as measured by CD3ζ chain expression in bystander T cells even in the presence of high H2O2 concentrations. Bystander NK cells, otherwise ROS sensitive, efficiently eliminate their K562 target cells under H2O2-induced oxidative stress when admixed with CAR-CAT T cells. This approach represents a novel means for protecting tumor-infiltrating cells from tumor-associated oxidative stress-mediated repression.


Asunto(s)
Catalasa/inmunología , Inmunoterapia Adoptiva/métodos , Células Asesinas Naturales/inmunología , Estrés Oxidativo/fisiología , Linfocitos T/inmunología , Western Blotting , Efecto Espectador/inmunología , Línea Celular , Separación Celular , Humanos , Receptores de Antígenos de Linfocitos T/inmunología , Proteínas Recombinantes de Fusión/inmunología , Transfección
5.
Cancer Cell ; 42(4): 623-645.e10, 2024 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-38490212

RESUMEN

Genes limiting T cell antitumor activity may serve as therapeutic targets. It has not been systematically studied whether there are regulators that uniquely or broadly contribute to T cell fitness. We perform genome-scale CRISPR-Cas9 knockout screens in primary CD8 T cells to uncover genes negatively impacting fitness upon three modes of stimulation: (1) intense, triggering activation-induced cell death (AICD); (2) acute, triggering expansion; (3) chronic, causing dysfunction. Besides established regulators, we uncover genes controlling T cell fitness either specifically or commonly upon differential stimulation. Dap5 ablation, ranking highly in all three screens, increases translation while enhancing tumor killing. Loss of Icam1-mediated homotypic T cell clustering amplifies cell expansion and effector functions after both acute and intense stimulation. Lastly, Ctbp1 inactivation induces functional T cell persistence exclusively upon chronic stimulation. Our results functionally annotate fitness regulators based on their unique or shared contribution to traits limiting T cell antitumor activity.


Asunto(s)
Sistemas CRISPR-Cas , Neoplasias , Humanos , Linfocitos T CD8-positivos , Neoplasias/genética
6.
Melanoma Res ; 33(1): 12-26, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36545919

RESUMEN

While immunotherapy has become standard-of-care for cutaneous melanoma patients, primary and acquired resistance prevent long-term benefits for about half of the late-stage patients. Pre-clinical models are essential to increase our understanding of the resistance mechanisms of melanomas, aiming to improve the efficacy of immunotherapy. Here, we present two novel syngeneic transplantable murine melanoma cell lines derived from the same primary tumor induced on BrafV600E Pten-/- mice: MeVa2.1 and MeVa2.2. Derivatives of these cell lines expressing the foreign antigen ovalbumin (dOVA) showed contrasting immune-mediated tumor control. MeVa2.2.dOVA melanomas were initially controlled in immune-competent hosts until variants grew out that had lost their antigens. By contrast, MeVa2.1.dOVA tumors were not controlled despite presenting the strong OVA antigen, as well as infiltration of tumor-reactive CD8+ T cells. MeVa2.1.dOVA displayed reduced sensitivity to T cell-mediated killing and growth inhibition in vitro by both IFN-γ and TNF-α. MeVa2.1.dOVA tumors were transiently controlled in vivo by either targeted therapy, adoptive T cell transfer, regulatory T cell depletion, or immune checkpoint blockade. MeVa2.1.dOVA could thus become a valuable melanoma model to evaluate novel immunotherapy combinations aiming to overcome immune resistance mechanisms.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Ratones , Animales , Melanoma/patología , Neoplasias Cutáneas/genética , Inmunoterapia , Linfocitos T CD8-positivos , Línea Celular Tumoral , Antígenos
7.
Cancer Immunol Res ; 11(4): 450-465, 2023 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-36753604

RESUMEN

Cross-presentation of tumor antigens by dendritic cells (DC) is crucial to prime, stimulate and restimulate CD8+ T cells. This process is important in initiating and maintaining an antitumor response. Here, we show that the presence of conventional type 1 DCs (cDC1), a DC subtype that excels in cross-presentation, in the tumor correlated with response to neoadjuvant immune checkpoint blockade (ICB) in melanoma. This led us to hypothesize that patients failing to respond to ICB could benefit from enhanced cross-presentation of tumor antigens. We therefore established a cross-presentation assay to screen over 5,500 compounds for enhancers of DC cross-presentation using induced T-cell proliferation as the readout. We identified 145 enhancers, including AZD5582, an antagonist of inhibitor of apoptosis proteins (IAP) cIAP1, cIAP2, and XIAP. AZD5582 treatment led to DC activation of the noncanonical NF-kB pathway, enhanced antigen import from endolysosomes into the cytosol, and increased expression of genes involved in cross-presentation. Furthermore, it upregulated expression of CD80, CD86, MHC class II, CD70 and secretion of TNF by DCs. This enhanced DC activation and maturation program was observed also in tumor-bearing mice upon AZD5582 treatment, culminating in an increased frequency of systemic tumor antigen-specific CD8+ T cells. Our results merit further exploration of AZD5582 to increase antigen cross-presentation for improving the clinical benefit of ICB in patients who are unlikely to respond to ICB.


Asunto(s)
Reactividad Cruzada , Melanoma , Ratones , Animales , Células Dendríticas , Presentación de Antígeno , Antígenos de Neoplasias , Proteínas Inhibidoras de la Apoptosis/metabolismo , Proliferación Celular
8.
Mol Ther ; 19(3): 594-601, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21157438

RESUMEN

DNA vaccination is an attractive approach to induce antigen-specific cytotoxic CD8(+) T lymphocytes (CTLs), which can mediate protective antitumor immunity. The potency of DNA vaccines encoding weakly immunogenic tumor-associated antigens (TAAs) can be enhanced by codelivering gene-encoded adjuvants. Pattern recognition receptors (PRRs) that sense intracellular DNA could potentially be used to harness intrinsic immune-stimulating properties of plasmid DNA vaccines. Consequently, the cytosolic DNA sensor, DNA-dependent activator of interferon (IFN) regulatory factors (DAI), was used as a genetic adjuvant. In vivo electroporation (EP) of mice with a DAI-encoding plasmid (pDAI) promoted transcription of genes encoding type I IFNs, proinflammatory cytokines, and costimulatory molecules. Coimmunization with pDAI and antigen-encoding plasmids enhanced in vivo antigen-specific proliferation, and induction of effector and memory CTLs. Moreover, codelivery of pDAI effectively promoted CTL and CD4(+) Th1 responses to the TAA survivin. The DAI-enhanced CTL induction required nuclear factor κB (NF-κB) activation and type I IFN signaling, but did not involve the IFN regulatory factor 3 (IRF3). Codelivery of pDAI also increased CTL responses to the melanoma-associated antigen tyrosinase-related protein-2 (TRP2), enhanced tumor rejection and conferred long-term protection against B16 melanoma challenge. This study constitutes "proof-of-principle" validating the use of intracellular PRRs as genetic adjuvants to enhance DNA vaccine potency.


Asunto(s)
Adyuvantes Inmunológicos/genética , Glicoproteínas , Neoplasias/inmunología , Linfocitos T Citotóxicos/inmunología , Vacunas de ADN , Inmunidad Adaptativa/genética , Animales , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Glicoproteínas/inmunología , Inmunidad Innata/genética , Proteínas Inhibidoras de la Apoptosis/inmunología , Oxidorreductasas Intramoleculares/inmunología , Activación de Linfocitos/genética , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plásmidos/genética , Plásmidos/inmunología , Proteínas de Unión al ARN , Proteínas Represoras/inmunología , Análisis de Supervivencia , Survivin , Vacunas de ADN/inmunología
9.
Cell Rep Med ; 3(6): 100655, 2022 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-35688159

RESUMEN

Tumor escape mechanisms for immunotherapy include deficiencies in antigen presentation, diminishing adaptive CD8+ T cell antitumor activity. Although innate natural killer (NK) cells are triggered by loss of MHC class I, their response is often inadequate. To increase tumor susceptibility to both innate and adaptive immune elimination, we performed parallel genome-wide CRISPR-Cas9 knockout screens under NK and CD8+ T cell pressure. We identify all components, RNF31, RBCK1, and SHARPIN, of the linear ubiquitination chain assembly complex (LUBAC). Genetic and pharmacologic ablation of RNF31, an E3 ubiquitin ligase, strongly sensitizes cancer cells to NK and CD8+ T cell killing. This occurs in a tumor necrosis factor (TNF)-dependent manner, causing loss of A20 and non-canonical IKK complexes from TNF receptor complex I. A small-molecule RNF31 inhibitor sensitizes colon carcinoma organoids to TNF and greatly enhances bystander killing of MHC antigen-deficient tumor cells. These results merit exploration of RNF31 inhibition as a clinical pharmacological opportunity for immunotherapy-refractory cancers.


Asunto(s)
Escape del Tumor , Ubiquitina-Proteína Ligasas , Células Asesinas Naturales , Factor de Necrosis Tumoral alfa/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
10.
Nat Commun ; 13(1): 1923, 2022 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-35395848

RESUMEN

The cytokine IFNγ differentially impacts on tumors upon immune checkpoint blockade (ICB). Despite our understanding of downstream signaling events, less is known about regulation of its receptor (IFNγ-R1). With an unbiased genome-wide CRISPR/Cas9 screen for critical regulators of IFNγ-R1 cell surface abundance, we identify STUB1 as an E3 ubiquitin ligase for IFNγ-R1 in complex with its signal-relaying kinase JAK1. STUB1 mediates ubiquitination-dependent proteasomal degradation of IFNγ-R1/JAK1 complex through IFNγ-R1K285 and JAK1K249. Conversely, STUB1 inactivation amplifies IFNγ signaling, sensitizing tumor cells to cytotoxic T cells in vitro. This is corroborated by an anticorrelation between STUB1 expression and IFNγ response in ICB-treated patients. Consistent with the context-dependent effects of IFNγ in vivo, anti-PD-1 response is increased in heterogenous tumors comprising both wildtype and STUB1-deficient cells, but not full STUB1 knockout tumors. These results uncover STUB1 as a critical regulator of IFNγ-R1, and highlight the context-dependency of STUB1-regulated IFNγ signaling for ICB outcome.


Asunto(s)
Interferón gamma , Neoplasias , Receptores de Interferón , Ubiquitina-Proteína Ligasas , Humanos , Inhibidores de Puntos de Control Inmunológico , Interferón gamma/metabolismo , Neoplasias/inmunología , Receptores de Interferón/metabolismo , Transducción de Señal , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Receptor de Interferón gamma
11.
J Virol ; 84(20): 10877-87, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20686047

RESUMEN

Chikungunya virus (CHIKV) is an emerging human pathogen transmitted by mosquitoes. Like that of other alphaviruses, CHIKV replication causes general host shutoff, leading to severe cytopathicity in mammalian cells, and inhibits the ability of infected cells to respond to interferon (IFN). Recent research, however, suggests that alphaviruses may have additional mechanisms to circumvent the host's antiviral IFN response. Here we show that CHIKV replication is resistant to inhibition by interferon once RNA replication has been established and that CHIKV actively suppresses the antiviral IFN response by preventing IFN-induced gene expression. Both CHIKV infection and CHIKV replicon RNA replication efficiently blocked STAT1 phosphorylation and/or nuclear translocation in mammalian cells induced by either type I or type II IFN. Expression of individual CHIKV nonstructural proteins (nsPs) showed that nsP2 was a potent inhibitor of IFN-induced JAK-STAT signaling. In addition, mutations in CHIKV-nsP2 (P718S) and Sindbis virus (SINV)-nsP2 (P726S) that render alphavirus replicons noncytopathic significantly reduced JAK-STAT inhibition. This host shutoff-independent inhibition of IFN signaling by CHIKV is likely to have an important role in viral pathogenesis.


Asunto(s)
Virus Chikungunya/fisiología , Virus Chikungunya/patogenicidad , Proteínas no Estructurales Virales/fisiología , Sustitución de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , Virus Chikungunya/efectos de los fármacos , Virus Chikungunya/genética , Chlorocebus aethiops , Cricetinae , ADN Viral/genética , Genes Virales , Humanos , Interferón Tipo I/farmacología , Interferón gamma/farmacología , Quinasas Janus/antagonistas & inhibidores , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/fisiología , Mutación , Proteínas Recombinantes , Replicón , Factores de Transcripción STAT/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Virus Sindbis/genética , Células Vero , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética , Replicación Viral/efectos de los fármacos , Replicación Viral/fisiología
12.
Cancer Res ; 81(7): 1775-1787, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33531370

RESUMEN

Although immune checkpoint blockade (ICB) has shown remarkable clinical benefit in a subset of patients with melanoma and lung cancer, most patients experience no durable benefit. The receptor tyrosine kinase AXL is commonly implicated in therapy resistance and may serve as a marker for therapy-refractory tumors, for example in melanoma, as we previously demonstrated. Here, we show that enapotamab vedotin (EnaV), an antibody-drug conjugate targeting AXL, effectively targets tumors that display insensitivity to immunotherapy or tumor-specific T cells in several melanoma and lung cancer models. In addition to its direct tumor cell killing activity, EnaV treatment induced an inflammatory response and immunogenic cell death in tumor cells and promoted the induction of a memory-like phenotype in cytotoxic T cells. Combining EnaV with tumor-specific T cells proved superior to either treatment alone in models of melanoma and lung cancer and induced ICB benefit in models otherwise insensitive to anti-PD-1 treatment. Our findings indicate that targeting AXL-expressing, immunotherapy-resistant tumors with EnaV causes an immune-stimulating tumor microenvironment and enhances sensitivity to ICB, warranting further investigation of this treatment combination. SIGNIFICANCE: These findings show that targeting AXL-positive tumor fractions with an antibody-drug conjugate enhances antitumor immunity in several humanized tumor models of melanoma and lung cancer.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoconjugados/uso terapéutico , Neoplasias Pulmonares/terapia , Melanoma/terapia , Proteínas Proto-Oncogénicas/inmunología , Proteínas Tirosina Quinasas Receptoras/inmunología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Línea Celular Tumoral , Terapia Combinada , Resistencia a Antineoplásicos/inmunología , Sinergismo Farmacológico , Células HEK293 , Humanos , Inhibidores de Puntos de Control Inmunológico/administración & dosificación , Inmunoconjugados/administración & dosificación , Inmunoterapia , Neoplasias Pulmonares/patología , Masculino , Melanoma/patología , Ratones , Ratones Desnudos , Ratones Transgénicos , Terapia Molecular Dirigida/métodos , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto , Tirosina Quinasa del Receptor Axl
13.
Clin Cancer Res ; 27(19): 5389-5400, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34230026

RESUMEN

PURPOSE: Combining anti-PD-1 + anti-CTLA-4 immune-checkpoint blockade (ICB) shows improved patient benefit, but it is associated with severe immune-related adverse events and exceedingly high cost. Therefore, there is a dire need to predict which patients respond to monotherapy and which require combination ICB treatment. EXPERIMENTAL DESIGN: In patient-derived melanoma xenografts (PDX), human tumor microenvironment (TME) cells were swiftly replaced by murine cells upon transplantation. Using our XenofilteR deconvolution algorithm we curated human tumor cell RNA reads, which were subsequently subtracted in silico from bulk (tumor cell + TME) patients' melanoma RNA. This produced a purely tumor cell-intrinsic signature ("InTumor") and a signature comprising tumor cell-extrinsic RNA reads ("ExTumor"). RESULTS: We show that whereas the InTumor signature predicts response to anti-PD-1, the ExTumor predicts anti-CTLA-4 benefit. In PDX, InTumorLO, but not InTumorHI, tumors are effectively eliminated by cytotoxic T cells. When used in conjunction, the InTumor and ExTumor signatures identify not only patients who have a substantially higher chance of responding to combination treatment than to either monotherapy, but also those who are likely to benefit little from anti-CTLA-4 on top of anti-PD-1. CONCLUSIONS: These signatures may be exploited to distinguish melanoma patients who need combination ICB blockade from those who likely benefit from either monotherapy.


Asunto(s)
Melanoma , Receptor de Muerte Celular Programada 1 , Animales , Antígeno CTLA-4 , Humanos , Inhibidores de Puntos de Control Inmunológico , Melanoma/tratamiento farmacológico , Melanoma/genética , Ratones , Receptor de Muerte Celular Programada 1/uso terapéutico , ARN , Microambiente Tumoral
14.
Nat Commun ; 11(1): 3946, 2020 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-32770055

RESUMEN

Melanomas can switch to a dedifferentiated cell state upon exposure to cytotoxic T cells. However, it is unclear whether such tumor cells pre-exist in patients and whether they can be resensitized to immunotherapy. Here, we chronically expose (patient-derived) melanoma cell lines to differentiation antigen-specific cytotoxic T cells and observe strong enrichment of a pre-existing NGFRhi population. These fractions are refractory also to T cells recognizing non-differentiation antigens, as well as to BRAF + MEK inhibitors. NGFRhi cells induce the neurotrophic factor BDNF, which contributes to T cell resistance, as does NGFR. In melanoma patients, a tumor-intrinsic NGFR signature predicts anti-PD-1 therapy resistance, and NGFRhi tumor fractions are associated with immune exclusion. Lastly, pharmacologic NGFR inhibition restores tumor sensitivity to T cell attack in vitro and in melanoma xenografts. These findings demonstrate the existence of a stable and pre-existing NGFRhi multitherapy-refractory melanoma subpopulation, which ought to be eliminated to revert intrinsic resistance to immunotherapeutic intervention.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Melanoma/tratamiento farmacológico , Proteínas del Tejido Nervioso/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Receptores de Factor de Crecimiento Nervioso/metabolismo , Neoplasias Cutáneas/tratamiento farmacológico , Linfocitos T Citotóxicos/inmunología , Animales , Antineoplásicos Inmunológicos/uso terapéutico , Factor Neurotrófico Derivado del Encéfalo/antagonistas & inhibidores , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Masculino , Melanoma/genética , Melanoma/inmunología , Melanoma/patología , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , RNA-Seq , Receptores de Factor de Crecimiento Nervioso/antagonistas & inhibidores , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Linfocitos T Citotóxicos/metabolismo , Escape del Tumor/genética , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Front Immunol ; 10: 2766, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31921104

RESUMEN

Tumor-infiltrating lymphocytes (TIL) are considered enriched for T cells recognizing shared tumor antigens or mutation-derived neoepitopes. We performed exome sequencing and HLA-A*02:01 epitope prediction from tumor cell lines from two HLA-A2-positive melanoma patients whose TIL displayed strong tumor reactivity. The potential neoepitopes were screened for recognition using autologous TIL by immunological assays and presentation on tumor major histocompatibility complex class I (MHC-I) molecules by Poisson detection mass spectrometry (MS). TIL from the patients recognized 5/181 and 3/49 of the predicted neoepitopes, respectively. MS screening detected 3/181 neoepitopes on tumor MHC-I from the first patient but only one was also among those recognized by TIL. Consequently, TIL enriched for neoepitope specificity failed to recognize tumor cells, despite being activated by peptides. For the second patient, only after IFN-γ treatment of the tumor cells was one of 49 predicted neoepitopes detected by MS, and this coincided with recognition by TIL sorted for the same specificity. Importantly, specific T cells could be expanded from patient and donor peripheral blood mononuclear cells (PBMC) for all neoepitopes recognized by TIL and/or detected on tumor MHC-I. In summary, stimulating the appropriate inflammatory environment within tumors may promote neoepitope MHC presentation while expanding T cells in blood may circumvent lack of specific TIL. The discordance in detection between physical and functional methods revealed here can be rationalized and used to improve neoantigen-targeted T cell immunotherapy.


Asunto(s)
Epítopos de Linfocito T/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Antígenos Específicos del Melanoma/inmunología , Melanoma/inmunología , Adulto , Anciano , Alelos , Presentación de Antígeno , Línea Celular Tumoral , Citometría de Flujo , Antígeno HLA-A2/inmunología , Antígenos de Histocompatibilidad/inmunología , Humanos , Inflamación/inmunología , Masculino , Espectrometría de Masas , Antígenos Específicos del Melanoma/genética , Mutación , Biblioteca de Péptidos , Secuenciación del Exoma
16.
Cancer Immunol Res ; 6(11): 1417-1425, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30143536

RESUMEN

Metastatic breast cancer is a fatal disease that responds poorly to treatment. Cancer vaccines targeting antigens expressed by metastatic breast cancer cells and cancer stem cells could function as anticancer therapies. Cripto-1 is an oncofetal protein overexpressed in invasive breast cancer and cancer-initiating cells. In this study, we explored the potential of a Cripto-1-encoding DNA vaccine to target breast cancer in preclinical mouse models. BALB/c mice and BALB-neuT mice were treated with a DNA vaccine encoding mouse Cripto-1 (mCr-1). BALB/c mice were challenged with murine breast cancer 4T1 cells or TUBO spheres; BALB-neuT mice spontaneously developed breast cancer. Tumor growth was followed in all mouse models and lung metastases were evaluated. In vitro assays were performed to identify the immune response elicited by vaccination. Vaccination against mCr-1 reduced primary tumor growth in the 4T1 metastatic breast cancer model and reduced lung metastatic burden. In BALB-neuT mice, because the primary tumors are Cripto-1 negative, vaccination against mCr-1 did not affect primary tumors but did reduce lung metastatic burden. Spheroid-cultured TUBO cells, derived from a BALB/neuT primary tumor, develop a cancer stem cell-like phenotype and express mCr-1. We observed reduced tumor growth in vaccinated mice after challenge with TUBO spheres. Our data indicate that vaccination against Cripto-1 results in a protective immune response against mCr-1 expressing and metastasizing cells. Targeting Cripto-1 by vaccination holds promise as an immunotherapy for treatment of metastatic breast cancer. Cancer Immunol Res; 6(11); 1417-25. ©2018 AACR.


Asunto(s)
Vacunas contra el Cáncer/farmacología , Factor de Crecimiento Epidérmico/genética , Neoplasias Mamarias Experimentales/terapia , Glicoproteínas de Membrana/genética , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/efectos de los fármacos , Vacunas de ADN/farmacología , Animales , Línea Celular Tumoral , Factor de Crecimiento Epidérmico/inmunología , Femenino , Inmunidad Humoral , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Glicoproteínas de Membrana/inmunología , Ratones Endogámicos BALB C , Proteínas de Neoplasias/inmunología , Células Madre Neoplásicas/inmunología
19.
Nat Med ; 24(2): 203-212, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29334371

RESUMEN

Intratumor heterogeneity is a key factor contributing to therapeutic failure and, hence, cancer lethality. Heterogeneous tumors show partial therapy responses, allowing for the emergence of drug-resistant clones that often express high levels of the receptor tyrosine kinase AXL. In melanoma, AXL-high cells are resistant to MAPK pathway inhibitors, whereas AXL-low cells are sensitive to these inhibitors, rationalizing a differential therapeutic approach. We developed an antibody-drug conjugate, AXL-107-MMAE, comprising a human AXL antibody linked to the microtubule-disrupting agent monomethyl auristatin E. We found that AXL-107-MMAE, as a single agent, displayed potent in vivo anti-tumor activity in patient-derived xenografts, including melanoma, lung, pancreas and cervical cancer. By eliminating distinct populations in heterogeneous melanoma cell pools, AXL-107-MMAE and MAPK pathway inhibitors cooperatively inhibited tumor growth. Furthermore, by inducing AXL transcription, BRAF/MEK inhibitors potentiated the efficacy of AXL-107-MMAE. These findings provide proof of concept for the premise that rationalized combinatorial targeting of distinct populations in heterogeneous tumors may improve therapeutic effect, and merit clinical validation of AXL-107-MMAE in both treatment-naive and drug-resistant cancers in mono- or combination therapy.


Asunto(s)
Inmunoconjugados/farmacología , Melanoma/tratamiento farmacológico , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas/inmunología , Proteínas Tirosina Quinasas Receptoras/inmunología , Animales , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/inmunología , Heterogeneidad Genética/efectos de los fármacos , Humanos , Inmunoconjugados/inmunología , Melanoma/genética , Melanoma/inmunología , Melanoma/patología , Ratones , Oligopéptidos/química , Oligopéptidos/inmunología , Oligopéptidos/farmacología , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/inmunología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/inmunología , Proteínas Tirosina Quinasas Receptoras/química , Proteínas Tirosina Quinasas Receptoras/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Tirosina Quinasa del Receptor Axl
20.
J Immunother Cancer ; 5(1): 73, 2017 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-28923105

RESUMEN

BACKGROUND: Adoptive natural killer (NK) cell transfer is being increasingly used as cancer treatment. However, clinical responses have so far been limited to patients with hematological malignancies. A potential limiting factor in patients with solid tumors is defective homing of the infused NK cells to the tumor site. Chemokines regulate the migration of leukocytes expressing corresponding chemokine receptors. Various solid tumors, including renal cell carcinoma (RCC), readily secrete ligands for the chemokine receptor CXCR2. We hypothesize that infusion of NK cells expressing high levels of the CXCR2 chemokine receptor will result in increased influx of the transferred NK cells into tumors, and improved clinical outcome in patients with cancer. METHODS: Blood and tumor biopsies from 14 primary RCC patients were assessed by flow cytometry and chemokine analysis. Primary NK cells were transduced with human CXCR2 using a retroviral system. CXCR2 receptor functionality was determined by Calcium flux and NK cell migration was evaluated in transwell assays. RESULTS: We detected higher concentrations of CXCR2 ligands in tumors compared with plasma of RCC patients. In addition, CXCL5 levels correlated with the intratumoral infiltration of CXCR2-positive NK cells. However, tumor-infiltrating NK cells from RCC patients expressed lower CXCR2 compared with peripheral blood NK cells. Moreover, healthy donor NK cells rapidly lost their CXCR2 expression upon in vitro culture and expansion. Genetic modification of human primary NK cells to re-express CXCR2 improved their ability to specifically migrate along a chemokine gradient of recombinant CXCR2 ligands or RCC tumor supernatants compared with controls. The enhanced trafficking resulted in increased killing of target cells. In addition, while their functionality remained unchanged compared with control NK cells, CXCR2-transduced NK cells obtained increased adhesion properties and formed more conjugates with target cells. CONCLUSIONS: To increase the success of NK cell-based therapies of solid tumors, it is of great importance to promote their homing to the tumor site. In this study, we show that stable engineering of human primary NK cells to express a chemokine receptor thereby enhancing their migration is a promising strategy to improve anti-tumor responses following adoptive transfer of NK cells.


Asunto(s)
Carcinoma de Células Renales/terapia , Ingeniería Genética/métodos , Neoplasias Renales/terapia , Células Asesinas Naturales/citología , Receptores de Interleucina-8B/genética , Traslado Adoptivo , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Línea Celular Tumoral , Movimiento Celular , Células Cultivadas , Quimiocina CXCL5/metabolismo , Femenino , Humanos , Interleucina-8/metabolismo , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Células Asesinas Naturales/metabolismo , Masculino , Receptores de Interleucina-8B/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA