Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Biomed Pharmacother ; 175: 116665, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38701564

RESUMEN

Opioid receptor agonists are often used when cancer patients undergo surgery or analgesic treatment. As analgesics in clinical care, opioids can provide intraoperative or to chronic cancer pain relief. Immune function plays an important role in anti-cancer therapy, with cellular immunity, comprised principally of T-lymphocytes and natural killer cells, representing the primary anti-cancer immune response. However, it remains unclear whether immune function is further affected with the use of opioids in already immunocompromised cancer patients. This article provides a review of the effects of commonly used clinical opioids, including morphine, oxycodone, fentanyl and tramadol, on immune function in cancer patients. It provides a summary of current evidence regarding the immunomodulatory effects of opioids in the cancer setting and mechanisms underlying these interactions.


Asunto(s)
Analgésicos Opioides , Neoplasias , Humanos , Analgésicos Opioides/uso terapéutico , Analgésicos Opioides/farmacología , Neoplasias/inmunología , Neoplasias/tratamiento farmacológico , Dolor en Cáncer/tratamiento farmacológico , Dolor en Cáncer/inmunología , Animales , Linfocitos T/inmunología , Linfocitos T/efectos de los fármacos , Sistema Inmunológico/efectos de los fármacos
2.
Nat Commun ; 12(1): 4558, 2021 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-34315904

RESUMEN

Patients with advanced stage cancers frequently suffer from severe pain as a result of bone metastasis and bone destruction, for which there is no efficacious treatment. Here, using multiple mouse models of bone cancer, we report that agonists of the immune regulator STING (stimulator of interferon genes) confer remarkable protection against cancer pain, bone destruction, and local tumor burden. Repeated systemic administration of STING agonists robustly attenuates bone cancer-induced pain and improves locomotor function. Interestingly, STING agonists produce acute pain relief through direct neuronal modulation. Additionally, STING agonists protect against local bone destruction and reduce local tumor burden through modulation of osteoclast and immune cell function in the tumor microenvironment, providing long-term cancer pain relief. Finally, these in vivo effects are dependent on host-intrinsic STING and IFN-I signaling. Overall, STING activation provides unique advantages in controlling bone cancer pain through distinct and synergistic actions on nociceptors, immune cells, and osteoclasts.


Asunto(s)
Neoplasias Óseas/complicaciones , Dolor en Cáncer/etiología , Dolor en Cáncer/inmunología , Proteínas de la Membrana/metabolismo , Neuronas/metabolismo , Analgésicos/farmacología , Animales , Neoplasias Óseas/sangre , Dolor en Cáncer/sangre , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Fémur/diagnóstico por imagen , Fémur/efectos de los fármacos , Fémur/patología , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Proteínas de Homeodominio/metabolismo , Hiperalgesia/complicaciones , Interferones/sangre , Interferones/metabolismo , Masculino , Neoplasias Mamarias Animales/complicaciones , Proteínas de la Membrana/agonistas , Ratones Endogámicos C57BL , Metástasis de la Neoplasia , Neuronas/efectos de los fármacos , Nocicepción/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Osteoclastos/patología , Osteogénesis/efectos de los fármacos , Receptor de Interferón alfa y beta/metabolismo , Transducción de Señal/efectos de los fármacos , Carga Tumoral/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Xantonas/farmacología
3.
Prostate ; 80(10): 782-794, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32407603

RESUMEN

BACKGROUND: Over 70% to 85% of men with advanced prostate cancer (PCa) develop bone metastases characterized by severe bone pain and increased likelihood of bone fracture. These clinical features result in decreased quality of life and act as a predictor of higher mortality. Mechanistically, the skeletal pathologies such as osteolytic lesions and abnormal osteoblastic activity drive these symptoms. The role of immune cells in bone cancer pain remains understudied, here we sought to recapitulate this symptomology in a murine model. METHODS: The prostate cancer bone metastasis-induced pain model (CIBP) was established by transplanting a mouse prostate cancer cell line into the femur of immunocompetent mice. Pain development, gait dynamics, and the changes in emotional activities like depression and anxiety were evaluated. Animal tissues including femurs, dorsal root ganglion (DRG), and spinal cord were collected at killing and microcomputed tomography (µCT), histology/immunohistochemistry, and quantitative immunofluorescent analysis were performed. RESULTS: Mice receiving prostate cancer cells showed a significantly lower threshold for paw withdrawal responses induced by mechanical stimulation compared with their control counterparts. Zero maze and DigiGait analyses indicated reduced and aberrant movement associated emotional activity compared with sham control at 8-weeks postinjection. The µCT analysis showed osteolytic and osteoblastic changes and a 50% reduction of the trabecular volumes within the prostate cancer group. Neurologically we demonstrated, increased calcitonin gene-related peptide (CGRP) and neuronal p75NTR immune-reactivities in both the projected terminals of the superficial dorsal horn and partial afferent neurons in DRG at L2 to L4 level in tumor-bearing mice. Furthermore, our data show elevated nerve growth factor (NGF) and TrkA immunoreactivities in the same segment of the superficial dorsal horn that were, however, not colocalized with CGRP and p75NTR . CONCLUSIONS: This study describes a novel immunocompetent model of CIBP and demonstrates the contribution of NGF and p75NTR to chronic pain in bone metastasis.


Asunto(s)
Neoplasias Óseas/patología , Neoplasias Óseas/secundario , Dolor en Cáncer/patología , Neoplasias de la Próstata/patología , Animales , Conducta Animal , Neoplasias Óseas/inmunología , Neoplasias Óseas/metabolismo , Péptido Relacionado con Gen de Calcitonina/inmunología , Péptido Relacionado con Gen de Calcitonina/metabolismo , Dolor en Cáncer/inmunología , Dolor en Cáncer/metabolismo , Modelos Animales de Enfermedad , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Inmunocompetencia , Masculino , Ratones , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Neuronas/metabolismo , Neuronas/patología , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/metabolismo , Receptores de Factor de Crecimiento Nervioso/inmunología , Receptores de Factor de Crecimiento Nervioso/metabolismo
4.
Brain Behav Immun ; 79: 139-151, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30685532

RESUMEN

Increasing evidence suggests that T cells participate in the pathology of neuropathic pain, as well as the activation of microglia. However, whether T cells infiltrate into the spinal cord and contribute to the development of bone cancer pain (BCP) remains unknown. Here, we used a mouse model of BCP to show that numbers of T cells infiltrated into the spinal cord after sarcoma cell implantation with increased BCP, and most infiltrating T cells in the spinal cord were CD3+CD4+ T cells. Both Th17 and Treg subpopulations were analyzed by immunofluorescence. Treg cells in the spinal cord were transiently up-regulated, followed by an imbalance towards Th17 afterwards, and elevated IL-17/IL-17A levels were observed in both blood and spinal cord. Meanwhile, TGF-ß, IL-6, and IL-23, the factors which regulate Th17/Treg differentiation, increased their expressions during the development of BCP. Additionally, IL-17A receptor (IL-17AR) was found to be expressed on microglia, and the level of IL-17AR increased with activated microglia during BCP development. Furthermore, BCP was ameliorated when IL-17/IL-17A neutralizing antibodies were intrathecally injected, accompanied with inhibited Th17/Treg infiltration and suppressed microglial activation. In conclusion, T cells infiltrated into the spinal cord with the imbalance of Th17/Treg towards Th17 during the development of BCP, which could promote the microglial activation and further increased BCP, while neutralizing IL-17/IL-17A in the spinal cord could ameliorate BCP. Our results suggest that targeting the imbalanced Th17/Treg infiltration in the spinal cord could be a novel strategy for BCP therapy.


Asunto(s)
Dolor en Cáncer/inmunología , Dolor en Cáncer/fisiopatología , Médula Espinal/inmunología , Animales , Neoplasias Óseas/fisiopatología , Dolor en Cáncer/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Factores de Transcripción Forkhead/metabolismo , Interleucina-17/análisis , Interleucina-17/metabolismo , Activación de Linfocitos , Ratones , Microglía/inmunología , Microglía/metabolismo , Dolor/inmunología , Dolor/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Células Th17/inmunología , Células Th17/metabolismo
5.
Br J Pharmacol ; 175(14): 2726-2736, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-28593737

RESUMEN

In patients with cancer, opioids are principally used for the management of acute surgical and chronic cancer-related pain. However, opioids have many non-analgesic effects, including direct and indirect effects on cancer cells and on anti-tumour immunity (NK cells, macrophages and T-cells). Direct effects on immune cells are manifested via opioid and non-opioid toll-like receptors, whereas indirect effects are manifested via the sympathetic nervous system and hypothalamic-pituitary-adrenal axis. Opioids can also decrease/alter immune cell infiltration into the tumour micro-environment. Animal models have shown that this is not a class effect, in that morphine and fentanyl suppress NK cell cytotoxicity; buprenorphine does not affect NK cell cytotoxicity, whereas tramadol increases NK cell cytotoxicity, reducing metastasis. In healthy individuals, morphine suppresses and fentanyl enhances NK cell cytotoxicity. In patients undergoing surgery, fentanyl decreased and tramadol increased NK cell cytotoxicity; clinical outcomes were not determined. Meta-analyses of opioid-sparing surgical studies report an association between improved recurrence-free and/or overall survival with regional/neuraxial anaesthesia compared with systemic opioids. In patients receiving opioids for non-surgical cancer-related pain, morphine has variable effects on immunity; clinical outcomes were not assessed. Although there is a potential association between systemic opioid administration and shorter survival in cancer patients with a prognosis of months to years, studies have not been designed to primarily assess survival, as a consequence of which causality cannot be apportioned. Pain is immunosuppressive, so analgesia is important. Opioids for cancer-related pain will continue to be recommended until definitive data on the effects of opioids on clinical outcomes in specific patient groups becomes available. LINKED ARTICLES: This article is part of a themed section on Emerging Areas of Opioid Pharmacology. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.14/issuetoc.


Asunto(s)
Analgésicos Opioides/farmacología , Dolor en Cáncer/inmunología , Sistema Inmunológico/efectos de los fármacos , Neoplasias/inmunología , Dolor Postoperatorio/inmunología , Analgésicos Opioides/uso terapéutico , Animales , Dolor en Cáncer/tratamiento farmacológico , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Neoplasias/cirugía , Dolor Postoperatorio/tratamiento farmacológico
6.
Integr Cancer Ther ; 17(2): 401-410, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-28870114

RESUMEN

HYPOTHESIS: Electroacupuncture (EA) has been used as an alternative analgesic therapy for hundreds of years, yet its analgesic potency and therapeutic advantage against bone cancer pain (BCP) in comparison with morphine remains unclear. This study aimed to investigate the effects of EA on mechanical allodynia and cellular immunity of BCP rats, and to further explore the potential mechanism. METHODS: The BCP model was established by implanting Walker 256 mammary gland carcinoma cells into the left tibia of adult female Sprague-Dawley rats. EA (dilatational wave, 2/100 Hz, 0.5 mA-1mA-1.5 mA for 10 minutes each intensity) was applied bilaterally to Zusanli (ST 36) and Kunlun (BL 60) for 30 minutes. Both EA stimulation and morphine (10 mg/kg, intraperitoneally) was given once every other day. Naloxone (0.3 mg/kg, intraperitoneally) was injected at 30 minutes prior to EA. Mechanical allodynia were demonstrated by paw withdrawal thresholds (PWTs) which measured by dynamic plantar aesthesiometer. T cell proliferation, percentage of CD3+, CD4+ and CD8+ T lymphocytes in spleen as well as expression of interleukin-2 (IL-2) in plasma were detected by WST-8, flow cytometry, and enzyme-linked immunosorbent assay technique, respectively. RESULTS: An intratibial inoculation of Walker 256 mammary gland carcinoma cells significantly decreased PWTs to mechanical stimuli. EA stimulation alleviated mechanical allodynia in BCP rats, and the analgesic potency of EA was weaker than that of morphine. In contrast to morphine, EA stimulation of BCP rats increased splenic concanavalin A (Con A)-induced T cell proliferation and plasma IL-2 content, as well as increased the percentages of splenic CD3+CD4+ and CD3+CD8+ T cell subsets. Moreover, both the analgesic effect and the partial immunomodulation of EA were suppressed by an intraperitoneal injection of naloxone. CONCLUSION: EA could significantly alleviate BCP-induced mechanical allodynia. Although the analgesic effect of EA was weaker than that of morphine, EA had an immunomodulation effect on cellular immunity. Both analgesic and immunomodulatory effect of EA might share the same mechanism via the opioid-mediated pathway, which needs further investigation.


Asunto(s)
Neoplasias Óseas/inmunología , Dolor en Cáncer/inmunología , Hiperalgesia/inmunología , Inmunidad Celular/inmunología , Animales , Complejo CD3/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Proliferación Celular/fisiología , Electroacupuntura/métodos , Femenino , Humanos , Ratas , Ratas Sprague-Dawley
7.
Acta Pharmacol Sin ; 37(6): 753-62, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27157092

RESUMEN

AIM: To investigate the mechanisms underlying the anti-nociceptive effect of minocycline on bone cancer pain (BCP) in rats. METHODS: A rat model of BCP was established by inoculating Walker 256 mammary carcinoma cells into tibial medullary canal. Two weeks later, the rats were injected with minocycline (50, 100 µg, intrathecally; or 40, 80 mg/kg, ip) twice daily for 3 consecutive days. Mechanical paw withdrawal threshold (PWT) was used to assess pain behavior. After the rats were euthanized, spinal cords were harvested for immunoblotting analyses. The effects of minocycline on NF-κB activation were also examined in primary rat astrocytes stimulated with IL-1ß in vitro. RESULTS: BCP rats had marked bone destruction, and showed mechanical tactile allodynia on d 7 and d 14 after the operation. Intrathecal injection of minocycline (100 µg) or intraperitoneal injection of minocycline (80 mg/kg) reversed BCP-induced mechanical tactile allodynia. Furthermore, intraperitoneal injection of minocycline (80 mg/kg) reversed BCP-induced upregulation of GFAP (astrocyte marker) and PSD95 in spinal cord. Moreover, intraperitoneal injection of minocycline (80 mg/kg) reversed BCP-induced upregulation of NF-κB, p-IKKα and IκBα in spinal cord. In IL-1ß-stimulated primary rat astrocytes, pretreatment with minocycline (75, 100 µmol/L) significantly inhibited the translocation of NF-κB to nucleus. CONCLUSION: Minocycline effectively alleviates BCP by inhibiting the NF-κB signaling pathway in spinal astrocytes.


Asunto(s)
Antibacterianos/uso terapéutico , Astrocitos/efectos de los fármacos , Neoplasias Óseas/complicaciones , Dolor en Cáncer/tratamiento farmacológico , Minociclina/uso terapéutico , FN-kappa B/inmunología , Médula Espinal/efectos de los fármacos , Analgésicos/uso terapéutico , Animales , Astrocitos/inmunología , Astrocitos/patología , Neoplasias Óseas/inmunología , Neoplasias Óseas/patología , Dolor en Cáncer/complicaciones , Dolor en Cáncer/inmunología , Dolor en Cáncer/patología , Línea Celular Tumoral , Femenino , Hiperalgesia/complicaciones , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/inmunología , Hiperalgesia/patología , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Médula Espinal/citología , Médula Espinal/inmunología , Médula Espinal/patología
8.
Curr Opin Support Palliat Care ; 10(2): 109-18, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26990052

RESUMEN

PURPOSE OF REVIEW: Most cancer patients experience pain and many will require opioids. However, the effects of opioids on cancer progression, metastasis, and recurrence is increasingly being questioned. There is evidence that opioids affect immune system function, angiogenesis, apoptosis, and invasion in a potentially deleterious manner. This review will examine the preclinical and clinical evidence. RECENT FINDINGS: Recent clinical data have struggled to find robust evidence that opioids promote cancer progression. Although most study has involved morphine, differential effects of other opioids on immune function and cancer are revealing a more complex picture. SUMMARY: Although there is a biologically plausible story, evidence for the action of opioids on cancer is mixed. Indeed, it may even be that in the chronic setting morphine has a beneficial effect on outcome in certain cancer types. This review critically examines and evaluates the evidence for the action of opioids on the processes involved in cancer progression. In the light of the uncertainty of opioid effect on cancer, any decision making should be tempered by knowing that stress and pain undoubtedly contribute to cancer progression.


Asunto(s)
Analgésicos Opioides/farmacología , Analgésicos Opioides/uso terapéutico , Dolor en Cáncer/tratamiento farmacológico , Dolor en Cáncer/fisiopatología , Sistema Inmunológico/efectos de los fármacos , Apoptosis/efectos de los fármacos , Dolor en Cáncer/inmunología , Citocinas/efectos de los fármacos , Progresión de la Enfermedad , Humanos , Sistema Inmunológico/metabolismo , Invasividad Neoplásica/patología , Metástasis de la Neoplasia , Recurrencia Local de Neoplasia , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/fisiopatología , Receptores Opioides/metabolismo , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...