Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Cancer Immunol Immunother ; 69(2): 255-261, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31781842

RESUMEN

A major challenge of cancer immunotherapy is the potential for undesirable effects on bystander cells and tumor-associated immune cells. Fundamentally, we need to understand what effect targeting tumor metabolism has upon the metabolism and phenotype of tumor-associated leukocytes, whose function can be critical for effective cancer therapeutic strategies. Undesirable effects of cancer therapeutics are a major reason for drug-associated toxicity, which confounds drug dosing and efficacy. As with any chemotherapeutic agent, drugs targeting tumor metabolism will exert potent effects on host stromal cells and tumor-associated leukocytes. Any drug targeting glycolysis, for example, could metabolically starve tumor-infiltrating T cells, inhibit their effector function and enable tumor progression. The targeting of oxidative phosphorylation in tumors will have complex effects on the polarization and function of tumor-associated macrophages. In short, we need to improve our understanding of tumor and immune cell metabolism and devise ways to specifically target tumors without compromising necessary host metabolism. Exploiting cell-specific metabolic pathways to directly target tumor cells may minimize detrimental effects on tumor-associated leukocytes.


Asunto(s)
Metabolismo Energético , Terapia Molecular Dirigida , Neoplasias/metabolismo , Neoplasias/terapia , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Biomarcadores , Comunicación Celular , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/efectos de la radiación , Humanos , Inmunomodulación , Leucocitos/efectos de los fármacos , Leucocitos/inmunología , Leucocitos/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Terapia Molecular Dirigida/métodos , Neoplasias/etiología , Neoplasias/patología , Transducción de Señal/efectos de los fármacos , Succinatos/metabolismo , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
2.
Cancer Immunol Immunother ; 69(2): 307-314, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31912230

RESUMEN

Sensory nerves sensitive to capsaicin are afferent nerve fibers which contain TRPV1 channels. Activation of these channels induces release of neuropeptides which regulate local blood flow and immune response. Inactivation of sensory neurons either with high-dose capsaicin treatment or local ablation of vagal sensory nerve activity markedly increases metastasis of breast carcinoma formed by 4T1 derivative cells. These cancer cells also induce an extensive systemic inflammatory response. Further findings have documented that lack of local sensory neuromediators alters phenotype of cancer cells within primary tumor leading to overgrowth of metastatic subsets. This might be due to decreases in local and systemic immune response to growing tumor. Specifically, Substance P, one of the most abundant sensory neuropeptides, enhances anti-tumoral immune response evoked by radiotherapy under in vivo conditions. These findings further suggest that activation of TRPV1 channels on sensory neurons may induce an anti-tumoral immune response. We are testing this hypothesis. Our initial results as reported here demonstrate anti-inflammatory consequences of low-dose systemic capsaicin treatment. In conclusion, sensory nerve fibers sensitive to capsaicin have important roles in defense against metastatic breast carcinoma; hence, controlled activation of these neural pathways might be effective in cancer therapy. Specifically, activation of sensory fibers of left vagus nerve using a perineuronal stimulation may inhibit metastasis of breast carcinoma. Likewise, pharmacological modulators of TRPV1 channels may induce anti-tumoral immune response. Exact players of this newly explored defense system are, however, only partly validated, and further studies are required.


Asunto(s)
Neoplasias de la Mama/etiología , Neoplasias de la Mama/metabolismo , Neuroinmunomodulación , Células Receptoras Sensoriales/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Desnervación , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Metástasis de la Neoplasia , Fenotipo , Radioterapia
3.
Cancer Immunol Immunother ; 69(2): 199-213, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31982939

RESUMEN

Neutrophils play a major role in tumor biology. Among other functions, neutrophils can release extracellular traps (NETs), mesh-like structures of decondensed chromatin fibers, in a process termed NETosis. Originally characterized as an antimicrobial mechanism, NETosis has been described in cancer, but cancer-related predisposition is not clear. In the current study, we investigated the predisposition of circulating neutrophils to release NETs in lung cancer and the impact of G-CSF on this function, comparing circulating neutrophils isolated from cancer patients to the LLC and AB12 mouse models. We find that neutrophils from both healthy donors and cancer patients display high NETotic potential, with 30-60% of cells undergoing NETosis upon PMA stimulation. In contrast, neutrophils isolated from tumor-bearing mice displayed only 4-5% NETotic cells, though significantly higher than naive controls (1-2%). Despite differential mechanisms of activation described, Ionomycin and PMA mainly triggered suicidal rather than vital NETosis. G-CSF secreting tumors did not increase NETotic rates in murine neutrophils, and direct G-CSF stimulation did not promote their NET release. In contrast, human neutrophils strongly responded to G-CSF stimulation resulting also in a higher response to PMA + G-CSF stimulation. Our data show clear differences in NETotic potentials between human and murine neutrophils. We do not find a predisposition of neutrophils to release NETs in lung cancer patients compared to healthy controls, whereas cancer may modulate neutrophils' NETotic potential in mice. G-CSF secreted from tumors differentially affects murine and human NETosis in cancer. These important differences should be considered in future studies of NETosis in cancer.


Asunto(s)
Trampas Extracelulares/fisiología , Neoplasias Pulmonares/inmunología , Neutrófilos/fisiología , Animales , Línea Celular Tumoral , Trampas Extracelulares/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos/farmacología , Humanos , Ionomicina/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Acetato de Tetradecanoilforbol/farmacología
4.
Cancer Immunol Immunother ; 69(2): 315-324, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31915854

RESUMEN

There is mounting evidence that the immune system can spontaneously clear malignant lesions before they manifest as overt cancer, albeit this activity has been difficult to demonstrate in humans. The calreticulin (CALR) exon 9 mutations are driver mutations in patients with chronic myeloproliferative neoplasms (MPN), which are chronic blood cancers. The CALR mutations generate a neo-antigen that is recognized by patient T cells, and T cells isolated from a patient with a CALR-mutation can recognize and kill autologous CALR-mutant cells. Surprisingly, healthy individuals display frequent and strong T cell responses to the CALR neo-antigens too. Furthermore, healthy individuals display immune responses to all parts of the mutant CALR epitope, and the CALR neo-epitope specific responses are memory T cell responses. These data suggest that although healthy individuals might acquire a CALR mutation, the mutant cells can be eliminated by the immune system. Additionally, a small fraction of healthy individuals harbor a CALR exon 9 mutation. Four healthy individuals carrying CALR mutations underwent a full medical examination including a bone marrow biopsy after a median follow up of 6.2 years. None of these patients displayed any signs of CALR-mutant MPN. Additionally, all healthy individuals displayed strong CALR neo-epitope specific T cell responses suggesting that these healthy individuals retained their CALR-mutant cells in the editing stage for several years. Thus, we suggest that CALR-mutant MPN could be a disease model of cancer immuno-editing, as we have demonstrated that CALR-mutant MPN displays all three stages described in the theory of cancer immuno-editing.


Asunto(s)
Susceptibilidad a Enfermedades , Neoplasias Hematológicas/etiología , Inmunomodulación , Escape del Tumor , Animales , Antígenos de Neoplasias/inmunología , Biomarcadores de Tumor , Calreticulina/genética , Calreticulina/metabolismo , Transformación Celular Neoplásica , Modelos Animales de Enfermedad , Epítopos/inmunología , Neoplasias Hematológicas/metabolismo , Humanos , Inmunomodulación/genética , Mutación , Escape del Tumor/genética
5.
Cancer Immunol Immunother ; 69(2): 293-306, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31953578

RESUMEN

Cancer immunotherapies are promising treatments for many forms of cancer. Nevertheless, the response rates to, e.g., immune checkpoint inhibitors (ICI), are still in low double-digit percentage. This calls for further therapy optimization that should take into account combination of immunotherapies with classical tumor therapies such as radiotherapy. By designing multimodal approaches, immune modulatory properties of certain radiation schemes, additional immune modulation by immunotherapy with ICI and hyperthermia, as well as patient stratification based on genetic and immune constitutions have to be considered. In this context, both the tumor and its microenvironment including cells of the innate and adaptive immune system have to be viewed in synopsis. Knowledge of immune activation and immune suppression by radiation is the basis for well-elaborated addition of certain immunotherapies. In this review, the focus is set on additional immune stimulation by hyperthermia and restoration of an immune response by ICI. The impact of radiation dose and fractionation on immune modulation in multimodal settings has to be considered, as the dynamics of the immune response and the timing between radiotherapy and immunotherapy. Another big challenge is the patient stratification that should be based on matrices of biomarkers, taking into account genetics, proteomics, radiomics, and "immunomics". One key aim is to turn immunological "cold" tumors into "hot" tumors, and to eliminate barriers of immune-suppressed or immune-excluded tumors. Comprehensive knowledge of immune alterations induced by radiation and immunotherapy when being applied together should be utilized for patient-adapted treatment planning and testing of innovative tumor therapies within clinical trials.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Diseño de Fármacos , Inmunomodulación/efectos de los fármacos , Neoplasias/etiología , Neoplasias/terapia , Animales , Antineoplásicos Inmunológicos/farmacología , Biomarcadores de Tumor , Terapia Combinada , Humanos , Hipertermia Inducida/métodos , Inmunidad , Factores Inmunológicos/farmacología , Inmunomodulación/efectos de la radiación , Inmunoterapia , Neoplasias/patología , Proyectos de Investigación , Resultado del Tratamiento , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Microambiente Tumoral/efectos de la radiación
6.
Cancer Immunol Immunother ; 69(2): 237-244, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31664482

RESUMEN

The TAM receptors-TYRO3, AXL, MERTK-are pleiotropically expressed receptors in both healthy and diseased tissue. A complex of the ligands Protein S (PROS1) or Growth Arrest-Specific 6 (GAS6) with apoptotic phosphatidylserine activates the TAM receptors. Hence, this receptor family is essential for the efferocytosis of apoptotic material by antigen-presenting cells. In addition, TAM receptors are expressed by virtually all cells of the tumor microenvironment. They are also potent oncogenes, frequently overexpressed in cancer and involved in survival and therapy resistance. Due to their pro-oncogenic and immune-inhibitory traits, TAM receptors have emerged as promising targets for cancer therapy. Recently, TAM receptors have been described to function as costimulatory molecules on human T cells. TAM receptors' ambivalent functions on many different cell types therefore make therapeutic targeting not straight-forward. In this review we summarize our current knowledge of the function of TAM receptors in the tumor microenvironment. We place particular focus on TAM receptors and the recently unraveled role of MERTK in activated T cells and potential consequences for anti-tumor immunity.


Asunto(s)
Neoplasias/etiología , Neoplasias/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Microambiente Tumoral/inmunología , Tirosina Quinasa c-Mer/metabolismo , Animales , Antineoplásicos/farmacología , Biomarcadores , Humanos , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Transducción de Señal , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética , Tirosina Quinasa c-Mer/genética , Tirosina Quinasa del Receptor Axl
7.
Cancer Immunol Immunother ; 69(2): 245-253, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31676924

RESUMEN

Schwann cells are the principal glial cells of the peripheral nervous system which maintain neuronal homeostasis. Schwann cells support peripheral nerve functions and play a critical role in many pathological processes including injury-induced nerve repair, neurodegenerative diseases, infections, neuropathic pain and cancer. Schwann cells are implicated in a wide range of diseases due, in part, to their ability to interact and modulate immune cells. We discuss the accumulating examples of how Schwann cell regulation of the immune system initiates and facilitates the progression of various diseases. Furthermore, we highlight how Schwann cells may orchestrate an immunosuppressive tumor microenvironment by polarizing and modulating the activity of the dendritic cells.


Asunto(s)
Susceptibilidad a Enfermedades , Inmunomodulación , Células de Schwann/inmunología , Células de Schwann/metabolismo , Animales , Biomarcadores , Humanos , Vaina de Mielina/inmunología , Vaina de Mielina/metabolismo , Transducción de Señal
8.
Cancer Immunol Immunother ; 69(2): 223-235, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31650200

RESUMEN

The lysyl oxidases (LOXs) are a family of enzymes deputed to cross-link collagen and elastin, shaping the structure and strength of the extracellular matrix (ECM). However, many novel "non-canonical" functions, alternative substrates, and regulatory mechanisms have been described and are being continuously elucidated. The activity of LOXs, therefore, appears to be integrated into a complex network of signals regulating many cell functions, including survival/proliferation/differentiation. Among these signaling pathways, TGF-ß and PI3K/Akt/mTOR, in particular, cross-talk extensively with each other and with LOXs also initiating complex feedback loops which modulate the activity of LOXs and direct the remodeling of the ECM. A growing body of evidence indicates that LOXs are not only important in the homeostasis of the normal structure of the ECM, but are also implicated in the establishment and maturation of the tumor microenvironment. LOXs' association with advanced and metastatic cancer is well established; however, there is enough evidence to support a significant role of LOXs in the transformation of normal epithelial cells, in the accelerated tumor development and the induction of invasion of the premalignant epithelium. A better understanding of LOXs and their interactions with the different elements of the tumor immune microenvironment will prove invaluable in the design of novel anti-tumor strategies.


Asunto(s)
Inmunidad , Neoplasias/etiología , Proteína-Lisina 6-Oxidasa/química , Proteína-Lisina 6-Oxidasa/inmunología , Microambiente Tumoral/inmunología , Animales , Biomarcadores , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Familia de Multigenes , Neoplasias/patología , Transducción de Señal , Relación Estructura-Actividad
9.
Cancer Immunol Immunother ; 69(2): 285-292, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31897662

RESUMEN

The wide-ranging collection of malignancies arising at the upper aerodigestive tract is categorized as head and neck cancer (HNC), the sixth most prevalent cancer worldwide. Infection with human papillomavirus (HPV) or exposure to carcinogens is the leading causes of HPV+ and HPV- HNCs development, respectively. HPV+ and HPV- HNCs are different in clinical and molecular aspects. Specifically, HPV- HNCs tightly associate with missense mutants of the TP53 gene (encoding for the p53 protein), suggesting a central role for mutant p53 gain-of-function (GOF) in driving tumorigenesis. In contrast, in HPV + HNC, the sequence of TP53 typically remains intact, while the protein is degraded. In tumor cells, the status of the TP53 gene affects the cargo of secreted exosomes. In this review, we describe the accumulated knowledge regarding the involvement of exosomes and p53 on cellular interactions between HPV+ and HPV- HNC cells, and the surrounding tumor microenvironment (TME). Moreover, we envision how TP53 status may determine exosomes cargo in HNC, and, consequently, modify the TME. The potential roles of exosomes described herein are based on both our studies and the studies of others on mutant p53-derived exosomes. Specifically, we showed how exosomes are shed by cancer cells harboring mutant p53 communicate with tumor-associated macrophages in the colon as well as with cancer-associated fibroblasts in the lung, creating immunosuppressive conditions and promoting invasiveness. Altogether, exosomes in HNC in the context of TP53 status are understudied and extensive research is required to shed light on the biology of HPV+ and HPV- HNC.


Asunto(s)
Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Exosomas/metabolismo , Neoplasias de Cabeza y Cuello/etiología , Neoplasias de Cabeza y Cuello/metabolismo , Mutación , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Animales , Susceptibilidad a Enfermedades , Matriz Extracelular/metabolismo , Predisposición Genética a la Enfermedad , Neoplasias de Cabeza y Cuello/patología , Humanos
10.
Cancer Immunol Immunother ; 69(2): 275-283, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31813053

RESUMEN

Cathepsins are lysosomal peptidases involved in intracellular protein catabolism as well as in various other physiological and pathological processes. Several members of the family, most notably cathepsins B, S, K and L, are frequently overexpressed in cancer and have been associated with remodeling of the proteins of the extracellular matrix, a process leading to tumor cell migration, invasion and metastasis. In addition, lysosomal cathepsins play a role in innate and adaptive immunity, regulation of antigen presentation, Toll-like receptor signaling, cytokine secretion, apoptosis, autophagy, differentiation, migration and cytotoxicity. In cancer, the cells of innate immunity, such as myeloid cells, are often subverted to the regulatory immunosuppressive phenotype. Most studies indicate that lysosomal cathepsins reinforce the pro-tumoral activity of myeloid-derived suppressor cells and tumor-associated macrophages as well as of neutrophils. On the other hand, in cytotoxic natural killer cells, tumor cells suppress lysosomal peptidases in their activation of perforin and granzymes, thus diminishing their killing ability. With multifaceted actions, lysosomal peptidases constitute an important regulatory mechanism for fine-tuning the anti-tumor immune response.


Asunto(s)
Inmunidad Innata , Lisosomas/metabolismo , Neoplasias/etiología , Neoplasias/metabolismo , Péptido Hidrolasas/metabolismo , Animales , Biomarcadores , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Humanos , Inmunidad , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Neoplasias/patología , Transducción de Señal , Receptores Toll-Like/metabolismo
11.
Cancer Immunol Immunother ; 69(2): 263-273, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31820042

RESUMEN

Current therapeutic approaches in malignancy are often based on combination therapies, reflecting present understanding of the way different players act together in cancer. The cooperative activity of several elements can potentiate the pro-metastatic functions of the cancer cells and of the tumor microenvironment (TME), together leading to a more aggressive disease phenotype. The design of improved therapeutic modalities requires better identification of networks that act at specific cancer-related settings, and of the molecular mechanisms involved. Such studies will indicate if therapies that co-target several factors or their receptors, simultaneously, could apply. Also, by delineating the intracellular pathways that are activated under such cooperative activities, it will be possible to determine whether to inhibit one specific molecular route that is shared by the different partners, or alternatively, design modalities that jointly target intracellular components acting in concert. This Focused Research Review illuminates the therapeutic relevance of this research field by describing our published findings in breast cancer-related publications, which identified networks that are established by the pro-inflammatory/pro-metastatic cytokine TNFα. It describes the additive/synergistic activities of TNFα with other soluble factors residing at the TME (e.g., IL-1ß, TGFß1, estrogen, EGF), with intracellular components such as the Ras oncogene, and with the tumor-stroma contexture through the activation of molecular cascades (Notch). The roles of the p65 (NF-κB) pathway-acting alone or in intricate relationships with other intracellular mechanisms-are described, the "TNFα-based network" is discussed as a general paradigm in malignancy and its clinical implications in cancer therapy are addressed.


Asunto(s)
Neoplasias/metabolismo , Neoplasias/patología , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Biomarcadores , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Humanos , Modelos Biológicos , Neoplasias/etiología , Fenotipo , Factor de Necrosis Tumoral alfa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA