Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Nature ; 611(7935): 405-412, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36323780

RESUMEN

Solid tumours are innervated by nerve fibres that arise from the autonomic and sensory peripheral nervous systems1-5. Whether the neo-innervation of tumours by pain-initiating sensory neurons affects cancer immunosurveillance remains unclear. Here we show that melanoma cells interact with nociceptor neurons, leading to increases in their neurite outgrowth, responsiveness to noxious ligands and neuropeptide release. Calcitonin gene-related peptide (CGRP)-one such nociceptor-produced neuropeptide-directly increases the exhaustion of cytotoxic CD8+ T cells, which limits their capacity to eliminate melanoma. Genetic ablation of the TRPV1 lineage, local pharmacological silencing of nociceptors and antagonism of the CGRP receptor RAMP1 all reduced the exhaustion of tumour-infiltrating leukocytes and decreased the growth of tumours, nearly tripling the survival rate of mice that were inoculated with B16F10 melanoma cells. Conversely, CD8+ T cell exhaustion was rescued in sensory-neuron-depleted mice that were treated with local recombinant CGRP. As compared with wild-type CD8+ T cells, Ramp1-/- CD8+ T cells were protected against exhaustion when co-transplanted into tumour-bearing Rag1-deficient mice. Single-cell RNA sequencing of biopsies from patients with melanoma revealed that intratumoral RAMP1-expressing CD8+ T cells were more exhausted than their RAMP1-negative counterparts, whereas overexpression of RAMP1 correlated with a poorer clinical prognosis. Overall, our results suggest that reducing the release of CGRP from tumour-innervating nociceptors could be a strategy to improve anti-tumour immunity by eliminating the immunomodulatory effects of CGRP on cytotoxic CD8+ T cells.


Asunto(s)
Linfocitos T CD8-positivos , Melanoma , Nociceptores , Animales , Ratones , Péptido Relacionado con Gen de Calcitonina/metabolismo , Péptido Relacionado con Gen de Calcitonina/farmacología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Melanoma/inmunología , Melanoma/patología , Nociceptores/fisiología , Células Receptoras Sensoriales/metabolismo , Neuritas/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/patología , Tasa de Supervivencia , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Genes RAG-1/genética , Humanos , Biopsia , Pronóstico
2.
FASEB J ; 38(13): e23803, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38963404

RESUMEN

Cancer neuroscience is an emerging field of cancer biology focused on defining the interactions and relationships between the nervous system, developing malignancies, and their environments. Our previous work demonstrates that small extracellular vesicles (sEVs) released by head and neck squamous cell carcinomas (HNSCCs) recruit loco-regional nerves to the tumor. sEVs contain a diverse collection of biological cargo, including microRNAs (miRNAs). Here, we asked whether two genes commonly amplified in HNSCC, CCND1, and PIK3CA, impact the sEV miRNA cargo and, subsequently, sEV-mediated tumor innervation. To test this, we individually overexpressed these genes in a syngeneic murine HNSCC cell line, purified their sEVs, and tested their neurite outgrowth activity on dorsal root ganglia (DRG) neurons in vitro. sEVs purified from Ccnd1-overexpressing cells significantly increased neurite outgrowth of DRG compared to sEVs from parental or Pik3ca over-expressing cells. When implanted into C57BL/6 mice, Ccnd1 over-expressing tumor cells promoted significantly more tumor innervation in vivo. qPCR analysis of sEVs shows that increased expression of Ccnd1 altered the packaging of miRNAs (miR-15-5p, miR-17-5p, and miR-21-5p), many of which target transcripts important in regulating axonogenesis. These data indicate that genetic amplifications harbored by malignancies impose changes in sEV miRNA cargo, which can influence tumorc innervation.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I , Vesículas Extracelulares , Neoplasias de Cabeza y Cuello , Ratones Endogámicos C57BL , MicroARNs , Animales , MicroARNs/genética , MicroARNs/metabolismo , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/genética , Ratones , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Fosfatidilinositol 3-Quinasa Clase I/genética , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Línea Celular Tumoral , Ciclina D1/genética , Ciclina D1/metabolismo , Ganglios Espinales/metabolismo , Humanos , Amplificación de Genes , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/patología
3.
Brain Behav Immun ; 107: 296-304, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36323360

RESUMEN

Persistent fatigue is a debilitating side effect that impacts a significant proportion of cancer survivors for which there is not yet an FDA-approved treatment. While certainly a multi-factorial problem, persistent fatigue could be due, in part, to associations learned during treatment. Therefore, we sought to investigate the role of associative learning in the persistence of fatigue using a preclinical model of cancer survivorship. For this purpose, we used a murine model of human papilloma virus-related head and neck cancer paired with a curative regimen of cisplatin-based chemoradiation in male C57BL/6J mice. Fatigue-like behavior was assessed by measuring variations in voluntary wheel running using a longitudinal design. Treatment robustly decreased voluntary wheel running, and this effect persisted for more than a month posttreatment. However, when wheels were removed during treatment, to minimize treatment-related fatigue, mice showed a more rapid return to baseline running levels. We confirmed that the delayed recovery observed in mice with continual wheel access was not due to increased treatment-related toxicity, in fact running attenuated cisplatin-induced kidney toxicity. Finally, we demonstrated that re-exposure to a treatment-related olfactory cue acutely re-instated fatigue. These data provide the first demonstration that associative processes can modulate the persistence of cancer-related fatigue-like behavior.


Asunto(s)
Supervivientes de Cáncer , Neoplasias , Humanos , Masculino , Ratones , Animales , Ratones Endogámicos C57BL , Actividad Motora , Investigación
4.
Anesth Analg ; 132(4): 1156-1163, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33323783

RESUMEN

BACKGROUND: Pain is one of the first presenting symptoms in patients with head and neck cancer, who often develop chronic and debilitating pain as the disease progresses. Pain is also an important prognostic marker for survival. Unfortunately, patients rarely receive effective pain treatment due to our limited knowledge of the mechanisms underlying head and neck cancer pain (HNCP). Pain is often associated with neuroinflammation and particularly interleukin (IL)-1 signaling. The purpose of this study is to develop a novel syngeneic model of HNCP in immunocompetent mice to examine the contribution of IL-1 signaling. METHODS: Male C57BL/6 mice were injected with a murine model of human papillomavirus (HPV+)-induced oropharyngeal squamous cell carcinoma in their right hindlimb to induce tumor growth. Pain sensitivity was measured via von Frey filaments. Spontaneous pain was assessed via the facial grimace scale. IL-1ß was measured by quantifying gene expression via quantitative polymerase chain reaction (qPCR) and enzyme-linked immunosorbent assay (ELISA). RESULTS: Pain hypersensitivity and spontaneous pain develop quickly after the implantation of tumor cells, a time when tumor volume is still insignificant. Spinal and circulating IL-1ß levels are significantly elevated in tumor-bearing mice. Blocking IL-1 signaling either by intrathecal administration of interleukin-1 receptor antagonist (IL-1ra) or by genetic deletion (interleukin-1 receptor knockout [Il1r1-/-]) does not alleviate HNCP. CONCLUSIONS: We established the first syngeneic model of HNCP in immunocompetent mice. Unlike inflammatory or nerve-injured pain, HNCP is independent of IL-1 signaling. These findings challenge the common belief that pain results from tissue compression or IL-1 signaling in patients with head and neck cancer.


Asunto(s)
Dolor en Cáncer/etiología , Interleucina-1beta/metabolismo , Neoplasias Orofaríngeas/complicaciones , Médula Espinal/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/complicaciones , Animales , Conducta Animal , Dolor en Cáncer/metabolismo , Dolor en Cáncer/fisiopatología , Línea Celular Tumoral , Proteína Antagonista del Receptor de Interleucina 1/genética , Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias Orofaríngeas/metabolismo , Neoplasias Orofaríngeas/virología , Umbral del Dolor , Papillomaviridae/patogenicidad , Transducción de Señal , Médula Espinal/fisiopatología , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/virología
5.
Gynecol Oncol ; 154(1): 228-235, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31003747

RESUMEN

OBJECTIVE: Recently, our laboratory identified sensory innervation within head and neck squamous cell carcinomas (HNSCCs) and subsequently defined a mechanism whereby HNSCCs promote their own innervation via the release of exosomes that stimulate neurite outgrowth. Interestingly, we noted that exosomes from human papillomavirus (HPV)-positive cell lines were more effective at promoting neurite outgrowth than those from HPV-negative cell lines. As nearly all cervical tumors are HPV-positive, we hypothesized that these findings would extend to cervical cancer. METHODS: We use an in vitro assay with PC12 cells to quantify the axonogenic potential of cervical cancer exosomes. PC12 cells are treated with cancer-derived exosomes, stained with the pan-neuronal marker (ß-III tubulin) and the number of neurites quantified. To assess innervation in cervical cancer, we immunohistochemically stained cervical cancer patient samples for ß-III tubulin and TRPV1 (sensory marker) and compared the staining to normal cervix. RESULTS: Here, we show the presence of sensory nerves within human cervical tumors. Additionally, we show that exosomes derived from HPV-positive cervical cancer cell lines effectively stimulate neurite outgrowth. CONCLUSIONS: These data identify sensory nerves as components of the cervical cancer microenvironment and suggest that tumor- derived exosomes promote their recruitment.


Asunto(s)
Vías Aferentes/patología , Exosomas/patología , Neoplasias del Cuello Uterino/patología , Vías Aferentes/metabolismo , Animales , Cuello del Útero/inervación , Exosomas/metabolismo , Femenino , Células HeLa , Papillomavirus Humano 16/aislamiento & purificación , Humanos , Inmunohistoquímica , Neuritas/metabolismo , Neuritas/patología , Células PC12 , Ratas , Canales Catiónicos TRPV/metabolismo , Tubulina (Proteína)/metabolismo , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/virología
6.
Pain ; 165(3): 608-620, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-37678566

RESUMEN

ABSTRACT: Severe pain is often experienced by patients with head and neck cancer and is associated with a poor prognosis. Despite its frequency and severity, current treatments fail to adequately control cancer-associated pain because of our lack of mechanistic understanding. Although recent works have shed some light of the biology underlying pain in HPV-negative oral cancers, the mechanisms mediating pain in HPV+ cancers remain unknown. Cancer-derived small extracellular vesicles (cancer-sEVs) are well positioned to function as mediators of communication between cancer cells and neurons. Inhibition of cancer-sEV release attenuated pain in tumor-bearing mice. Injection of purified cancer-sEVs is sufficient to induce pain hypersensitivity in naive mice that is prevented by QX-314 treatment and in Trpv1-/- mice. Cancer-sEVs triggered calcium influx in nociceptors, and inhibition or ablation of nociceptors protects against cancer pain. Interrogation of published sequencing data of human sensory neurons exposed to human cancer-sEVs suggested a stimulation of protein translation in neurons. Induction of translation by cancer-sEVs was validated in our mouse model, and its inhibition alleviated cancer pain in mice. In summary, our work reveals that HPV+ head and neck squamous cell carcinoma-derived sEVs alter TRPV1+ neurons by promoting nascent translation to mediate cancer pain and identified several promising therapeutic targets to interfere with this pathway.


Asunto(s)
Dolor en Cáncer , Vesículas Extracelulares , Neoplasias de Cabeza y Cuello , Infecciones por Papillomavirus , Humanos , Animales , Ratones , Dolor en Cáncer/etiología , Neoplasias de Cabeza y Cuello/complicaciones , Dolor , Neuronas , Canales Catiónicos TRPV/genética
7.
bioRxiv ; 2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-37905135

RESUMEN

Cancer patients often experience changes in mental health, prompting an exploration into whether nerves infiltrating tumors contribute to these alterations by impacting brain functions. Using a male mouse model for head and neck cancer, we utilized neuronal tracing techniques and show that tumor-infiltrating nerves indeed connect to distinct brain areas via the ipsilateral trigeminal ganglion. The activation of this neuronal circuitry led to behavioral alterations represented by decreased nest-building, increased latency to eat a cookie, and reduced wheel running. Tumor-infiltrating nociceptor neurons exhibited heightened activity, as indicated by increased calcium mobilization. Correspondingly, the specific brain regions receiving these neural projections showed elevated cFos and delta FosB expression in tumor-bearing mice, alongside markedly intensified calcium responses compared to non-tumor-bearing counterparts. The genetic elimination of nociceptor neurons in tumor-bearing mice led to decreased brain Fos expression and mitigated the behavioral alterations induced by the presence of the tumor. While analgesic treatment successfully restored behaviors involving oral movements to normalcy in tumor-bearing mice, it did not have a similar therapeutic effect on voluntary wheel running. This discrepancy points towards an intricate relationship, where pain is not the exclusive driver of such behavioral shifts. Unraveling the interaction between the tumor, infiltrating nerves, and the brain is pivotal to developing targeted interventions to alleviate the mental health burdens associated with cancer. Significance Statement: Head and neck cancers are infiltrated by sensory nerves which connect to a pre-existing circuit that includes areas in the brain. Neurons within this circuit are altered and mediate modifications in behavior.

8.
bioRxiv ; 2024 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-39253487

RESUMEN

Nociceptor neurons impact tumor immunity. Removing nociceptor neurons reduced myeloid-derived suppressor cell (MDSCs) tumor infiltration in mouse models of head and neck carcinoma and melanoma. Carcinoma-released small extracellular vesicles (sEVs) attract nociceptive nerves to tumors. sEV-deficient tumors fail to develop in mice lacking nociceptor neurons. Exposure of dorsal root ganglia (DRG) neurons to cancer sEVs elevated expression of Substance P, IL-6 and injury-related neuronal markers while treatment with cancer sEVs and cytotoxic CD8 T-cells induced an immunosuppressive state (increased exhaustion ligands and cytokines). Cancer patient sEVs enhanced DRG responses to capsaicin, indicating increased nociceptor sensitivity. Conditioned media from DRG and cancer cell co-cultures promoted expression of MDSC markers in primary bone marrow cells while DRG conditioned media together with cancer sEVs induced checkpoint expression on T-cells. Our findings indicate that nociceptor neurons facilitate CD8+ T cell exhaustion and enhance MDSC infiltration. Targeting nociceptor-released IL-6 emerges as a novel strategy to disrupt harmful neuro-immune interactions in cancer and enhance anti-tumor immunity.

9.
Elife ; 132024 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-39302290

RESUMEN

Cancer patients often experience changes in mental health, prompting an exploration into whether nerves infiltrating tumors contribute to these alterations by impacting brain functions. Using a mouse model for head and neck cancer and neuronal tracing, we show that tumor-infiltrating nerves connect to distinct brain areas. The activation of this neuronal circuitry altered behaviors (decreased nest-building, increased latency to eat a cookie, and reduced wheel running). Tumor-infiltrating nociceptor neurons exhibited heightened calcium activity and brain regions receiving these neural projections showed elevated Fos as well as increased calcium responses compared to non-tumor-bearing counterparts. The genetic elimination of nociceptor neurons decreased brain Fos expression and mitigated the behavioral alterations induced by the presence of the tumor. While analgesic treatment restored nesting and cookie test behaviors, it did not fully restore voluntary wheel running indicating that pain is not the exclusive driver of such behavioral shifts. Unraveling the interaction between the tumor, infiltrating nerves, and the brain is pivotal to developing targeted interventions to alleviate the mental health burdens associated with cancer.


A lot of cancer survivors experience a decline in mental health, persisting often decades after successful treatment. Many factors contribute to this reduced mental well-being, including the physical, emotional and financial stresses they experience. Scientists think that the increased prevalence of mental health disorders among cancer patients and survivors may also be linked to the cancer itself. Previous research has shown that most tumors, in particular in melanomas, cervical and ovarian cancers, and head and neck cancers, contain sensory nerves that sense thermal, mechanical and chemical changes and so alert an organism about a potential danger, such as extreme temperature, pressure, changes in pH or inflammation. To investigate whether these nerves contribute to the worsened mental health of cancer patients, Barr, Walz et al. studied male mice with tumors growing in their mouths, mimicking the disease of patients with head and neck cancers. The mice with tumors exhibited several altered behaviors linked to their well-being, suggesting that they had reduced overall health compared to mice without tumors. For example, they were less inclined to build nests, accept treats or run on a wheel. Next, Barr, Walz et al. injected a fluorescent dye into the tumors to label the nerves inside the cancerous growths. Fluorescence microscopy and imaging studies revealed that, days later, the dye had traveled to multiple regions of the brain, indicating that the nerves in the tumors had connected to a preexisting nerve circuit that included these brain regions. Further experiments revealed that the nerve cells in these brain regions were more active in mice with tumors and had different functional properties compared to mice without tumors. Removing the connecting nerves either genetically or with a drug improved all the behaviors of the mice with tumors. Treating the mice with painkillers also improved some but not all of their behaviors, indicating that pain is not the exclusive driver of such behavioral shifts. These two experiments suggest that the nerves from the tumors relay information about pain to the brain and contribute to reduced well-being of the mice. Further studies will test whether these tumor-brain connections also contribute to behavioral changes in mice with other types of cancer. The data suggest that disrupting the neural connections between a tumor and the brain may improve the mental health of patients with cancer, but more research is needed to establish this link.


Asunto(s)
Encéfalo , Modelos Animales de Enfermedad , Neoplasias de Cabeza y Cuello , Animales , Ratones , Neoplasias de Cabeza y Cuello/fisiopatología , Conducta Animal , Neuronas/fisiología , Neuronas/metabolismo , Ratones Endogámicos C57BL , Masculino
10.
Cancer Discov ; 14(4): 669-673, 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38571430

RESUMEN

SUMMARY: The field of cancer neuroscience has begun to define the contributions of nerves to cancer initiation and progression; here, we highlight the future directions of basic and translational cancer neuroscience for malignancies arising outside of the central nervous system.


Asunto(s)
Neoplasias , Neurociencias , Humanos , Sistema Nervioso Central , Predicción , Proteómica
11.
Int J Cancer ; 133(1): 120-9, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23292955

RESUMEN

The increasing incidence of human papillomavirus (HPV) related oropharyngeal squamous cell carcinoma (OSSC) demands development of novel therapies. Despite presenting at a more advanced stage, HPV(+) oropharyngeal squamous cell carcinoma (OSCC) have a better prognosis than their HPV(-) counterparts. We have previously demonstrated that clearance of HPV(+) OSCC during treatment with radiation and chemotherapy requires an immune response which is likely responsible for the improved clinical outcomes. To further elucidate the mechanism of immune-mediated clearance, we asked whether radiation therapy induces tumor cell changes that allow the body to recognize and aid in tumor clearance. Here, we describe a radiation-induced change in tumor surface protein expression that is critical for immune-mediated clearance. Radiation therapy decreases surface expression of CD47, a self-marker. CD47 is frequently overexpressed in head and neck squamous cell carcinoma and radiation induces a decrease of CD47 in a dose-dependent manner. We show that both in vitro and in vivo tumor cell CD47 protein levels are restored over time after sublethal radiation exposure and that protein levels on adjacent, normal tissues remain unaffected. Furthermore, reduction of tumor cell CD47 increases phagocytosis of these cells by dendritic cells and leads to increased interferon gamma and granzyme production from mixed lymphocytes. Finally, decreasing tumor cell CD47 in combination with standard radiation and chemotherapy results in improved immune-mediated tumor clearance in vivo. These findings help define an important mechanism of radiation-related immune clearance and suggest that decreasing CD47 specifically on tumor cells may be a good therapeutic target for HPV related disease.


Asunto(s)
Antígeno CD47/inmunología , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/radioterapia , Papillomavirus Humano 16 , Neoplasias Orofaríngeas/inmunología , Neoplasias Orofaríngeas/radioterapia , Infecciones por Papillomavirus/complicaciones , Animales , Antineoplásicos/uso terapéutico , Carcinoma de Células Escamosas/virología , Línea Celular Tumoral , Supervivencia Celular , Cisplatino/uso terapéutico , Relación Dosis-Respuesta en la Radiación , Granzimas/inmunología , Papillomavirus Humano 16/aislamiento & purificación , Humanos , Interferón gamma/inmunología , Masculino , Ratones , Neoplasias Orofaríngeas/virología , Fagocitosis , Radioterapia Adyuvante
12.
Sci Adv ; 9(19): eade4443, 2023 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-37163587

RESUMEN

The molecular and functional contributions of intratumoral nerves to disease remain largely unknown. We localized synaptic markers within tumors suggesting that these nerves form functional connections. Consistent with this, electrophysiological analysis shows that malignancies harbor significantly higher electrical activity than benign disease or normal tissues. We also demonstrate pharmacologic silencing of tumoral electrical activity. Tumors implanted in transgenic animals lacking nociceptor neurons show reduced electrical activity. These data suggest that intratumoral nerves remain functional at the tumor bed. Immunohistochemical staining demonstrates the presence of the neuropeptide, Substance P (SP), within the tumor space. We show that tumor cells express the SP receptor, NK1R, and that ligand/receptor engagement promotes cellular proliferation and migration. Our findings identify a mechanism whereby intratumoral nerves promote cancer progression.


Asunto(s)
Neoplasias de la Mama , Neuronas , Neoplasias Ováricas , Carcinoma de Células Escamosas de Cabeza y Cuello , Animales , Ratones , Modelos Animales de Enfermedad , Humanos , Neoplasias de la Mama/epidemiología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Sustancia P/metabolismo , Línea Celular Tumoral , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Carcinoma de Células Escamosas de Cabeza y Cuello/virología , Neoplasias Ováricas/epidemiología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/secundario , Neuronas/patología , Ratones Endogámicos C57BL , Organismos Libres de Patógenos Específicos , Ovario/inervación , Virus del Papiloma Humano , Análisis de Supervivencia
13.
FASEB Bioadv ; 4(1): 29-42, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35024571

RESUMEN

The identification of nerves in the tumor microenvironment has ushered in a new area of research in cancer biology. Numerous studies demonstrate the presence of various types of peripheral nerves (sympathetic, parasympathetic, sensory) within the tumor microenvironment; moreover, an increased density of nerves in the tumor microenvironment correlates with worse prognosis. In this review, we address the current understanding of nerve-mediated alterations of the tumor microenvironment and how they impact disease through a variety of processes, including direct nerve-cancer cell communication, alteration of the infiltrative immune population, and alteration of stromal components.

14.
Cancers (Basel) ; 14(10)2022 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-35626103

RESUMEN

BACKGROUND: Chemoradiotherapy is a standard treatment for HNSCC. Blockade of the PD-1/L1-2 interaction may represent a target to overcome immune escape during this treatment. METHODS: Utilizing a HNSCC mEERL C57BL/6 mouse model, we evaluated a PD-1 blockade alone or in combination with cisplatin-based chemoradiotherapy. Next, we evaluated peripheral blood mononuclear cells (PBMCs) with relative PD-1, TIM-3, and LAG-3 expression, and myeloid-derived suppressor-like (MDSC-like) populations from a clinical trial evaluating PD-1 blockade with chemoradiotherapy in HNSCC. Finally, we analyzed the effect of therapy on human T-cell clonality through T-cell Receptor (TCR) sequencing. RESULTS: Anti-PD-1 monotherapy induced no response in the mEERL model; however, combination with chemoradiotherapy improved tumor clearance and survival. PBMCs from patients treated with this combination therapy demonstrate a decline in circulating T-cell populations with knockdown of PD-1 expressing CD3+CD4+ and CD3+CD8+ T cells during treatment. However, TIM-3, LAG-3 expressing T-cell and MDSC-like populations concordantly rose. During treatment, the TCR repertoire demonstrates overall clonal expansion, with both unique and previously reported T-cell clones. CONCLUSIONS: Our murine HNSCC model demonstrates efficacy of PD-1 blockade during chemoradiotherapy. However, while PD-1-expressing T cells decreased with this therapy, human PBMC findings also identified an increase in populations contributing to immune exhaustion. These findings further characterize PD-1 blockade during chemoradiotherapy for HNSCC and highlight potential competing mechanisms of immune evasion.

15.
Cells ; 10(12)2021 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-34944001

RESUMEN

Dense tumor innervation is associated with enhanced cancer progression and poor prognosis. We observed innervation in breast, prostate, pancreatic, lung, liver, ovarian, and colon cancers. Defining innervation in high-grade serous ovarian carcinoma (HGSOC) was a focus since sensory innervation was observed whereas the normal tissue contains predominantly sympathetic input. The origin, specific nerve type, and the mechanisms promoting innervation and driving nerve-cancer cell communications in ovarian cancer remain largely unknown. The technique of neuro-tracing enhances the study of tumor innervation by offering a means for identification and mapping of nerve sources that may directly and indirectly affect the tumor microenvironment. Here, we establish a murine model of HGSOC and utilize image-guided microinjections of retrograde neuro-tracer to label tumor-infiltrating peripheral neurons, mapping their source and circuitry. We show that regional sensory neurons innervate HGSOC tumors. Interestingly, the axons within the tumor trace back to local dorsal root ganglia as well as jugular-nodose ganglia. Further manipulations of these tumor projecting neurons may define the neuronal contributions in tumor growth, invasion, metastasis, and responses to therapeutics.


Asunto(s)
Cistadenocarcinoma Seroso/patología , Tejido Nervioso/patología , Neoplasias Ováricas/patología , Animales , Cistadenocarcinoma Seroso/diagnóstico por imagen , Modelos Animales de Enfermedad , Femenino , Ganglios Espinales/metabolismo , Ratones Endogámicos C57BL , Tejido Nervioso/diagnóstico por imagen , Neoplasias Ováricas/diagnóstico por imagen , Fosfohidrolasa PTEN/metabolismo , Células Receptoras Sensoriales/patología , Proteína p53 Supresora de Tumor/metabolismo , Ultrasonografía
16.
Cancer Res ; 79(14): 3529-3535, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31088834

RESUMEN

The naïve view of tumors as isolated islands of rogue cells has given way to a deeper understanding of cancer as being closer to a foreign organ. This "organ" contains immunologic, vascular, and neural connections to its host that provide not only mechanisms for disease progression but also opportunities for therapeutic intervention. The presence of nerves within tumor tissues has long been appreciated. However, a mechanistic understanding of how tumors recruit nerves has been slower to emerge. Tumor release of neurotrophic factors and axonal guidance molecules likely directs axons toward the tumor bed. Newly emerging data support a contribution of tumor-released exosomes in the induction of axonogenesis toward the tumor. Exosomes, small membrane-bound vesicles that carry a complex cargo (DNA, RNA, miRNA, lipids, and proteins), protect their cargo from the low pH of the tumor microenvironment. They also represent an efficient means of local and distal communication between the tumor and potentially innervating nerves. Likely, a combination of neurotrophins, guidance molecules, and exosomes work in concert to promote tumor innervation. As such, defining the critical components driving tumor innervation will identify new targets for intervention. Moreover, with a deepening understanding, tumor innervation may emerge as a new hallmark of cancer.


Asunto(s)
Exosomas/patología , Neoplasias/patología , Sistema Nervioso/patología , Animales , Axones/metabolismo , Axones/patología , Exosomas/metabolismo , Humanos , Neoplasias/metabolismo , Sistema Nervioso/metabolismo , Microambiente Tumoral
17.
Int J Tryptophan Res ; 12: 1178646919872508, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31496720

RESUMEN

The expression of indoleamine 2,3 dioxygenase (IDO) by tumors can contribute to immunotolerance, and IDO induced by inflammation can also increase risk for the development of behavioral alterations. Thus, this study was initiated to determine whether IDO inhibition, intended to facilitate tumor clearance in response to treatment, attenuates behavioral alterations associated with tumor growth and treatment. We used a murine model of human papilloma virus-related head and neck cancer. We confirmed that tumor cells express IDO and expression was increased by radiotherapy. Interestingly, inhibition of IDO activation by the competitive inhibitor 1-methyl tryptophan mildly exacerbated treatment-associated burrowing deficits (burrowing is a sensitive index of sickness in tumor-bearing mice). Genetic deletion of IDO worsened tumor outcomes and had no effect on the behavioral response as by decreased burrowing or reduced voluntary wheel running. In contrast, oral administration of a specific inhibitor of IDO1 provided no apparent benefit on the tumor response to cancer therapy, yet decreased voluntary wheel-running activity independent of treatment. These results indicate that, independent of its potential effect on tumor clearance, inhibition of IDO does not improve cancer-related symptoms.

18.
Cancers (Basel) ; 10(12)2018 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-30513596

RESUMEN

Tumor cell metabolism differs from that of normal cells, conferring tumors with metabolic advantages but affording opportunities for therapeutic intervention. Accordingly, metabolism-targeting therapies have shown promise. However, drugs targeting singular metabolic pathways display limited efficacy, in part due to the tumor's ability to compensate by using other metabolic pathways to meet energy and growth demands. Thus, it is critical to identify novel combinations of metabolism-targeting drugs to improve therapeutic efficacy in the face of compensatory cellular response mechanisms. Our lab has previously identified that the anti-cancer activity of propranolol, a non-selective beta-blocker, is associated with inhibition of mitochondrial metabolism in head and neck squamous cell carcinoma (HNSCC). In response to propranolol, however, HNSCC exhibits heightened glycolytic activity, which may limit the effectiveness of propranolol as a single agent. Thus, we hypothesized that propranolol's metabolic effects promote a state of enhanced glucose dependence, and that propranolol together with glycolytic inhibition would provide a highly effective therapeutic combination in HNSCC. Here, we show that glucose deprivation synergizes with propranolol for anti-cancer activity, and that the rational combination of propranolol and dichloroacetate (DCA), a clinically available glycolytic inhibitor, dramatically attenuates tumor cell metabolism and mTOR signaling, inhibits proliferation and colony formation, and induces apoptosis. This therapeutic combination displays efficacy in both human papillomavirus-positive HPV(+) and HPV(-) HNSCC cell lines, as well as a recurrent/metastatic model, while leaving normal tonsil epithelial cells relatively unaffected. Importantly, the combination significantly delays tumor growth in vivo with no evidence of toxicity. Additionally, the combination of propranolol and DCA enhances the effects of chemoradiation and sensitizes resistant cells to cisplatin and radiation. This novel therapeutic combination represents a promising treatment strategy which may overcome some of the limitations of targeting individual metabolic pathways in cancer.

19.
Cancer Res ; 78(3): 695-705, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29217760

RESUMEN

Fatigue is the most common symptom of cancer at diagnosis, yet causes and effective treatments remain elusive. As tumors can be highly inflammatory, it is generally accepted that inflammation mediates cancer-related fatigue. However, evidence to support this assertion is mostly correlational. In this study, we directly tested the hypothesis that fatigue results from propagation of tumor-induced inflammation to the brain and activation of the central proinflammatory cytokine, IL1. The heterotopic syngeneic murine head and neck cancer model (mEER) caused systemic inflammation and increased expression of Il1b in the brain while inducing fatigue-like behaviors characterized by decreased voluntary wheel running and exploratory activity. Expression of Il1b in the brain was not associated with any alterations in motivation, measured by responding in a progressive ratio schedule of food reinforcement, depression-like behaviors, or energy balance. Decreased wheel running occurred prior to Il1b detection in the brain, when systemic inflammation was minimal. Furthermore, mice null for two components of IL1ß signaling, the type 1 IL1 receptor or the receptor adapter protein MyD88, were not protected from tumor-induced decreases in wheel running, despite attenuated cytokine action and expression. Behavioral and inflammatory analysis of four additional syngeneic tumor models revealed that tumors can induce fatigue regardless of their systemic or central nervous system inflammatory potential. Together, our results show that brain IL1 signaling is not necessary for tumor-related fatigue, dissociating this type of cancer sequela from systemic cytokine expression.Significance: These findings challenge the current understanding of fatigue in cancer patients, the most common and debilitating sequela associated with malignancy. Cancer Res; 78(3); 695-705. ©2017 AACR.


Asunto(s)
Encéfalo/patología , Fatiga/etiología , Neoplasias de Cabeza y Cuello/complicaciones , Inflamación/etiología , Interleucina-1beta/metabolismo , Factor 88 de Diferenciación Mieloide/fisiología , Receptores Tipo I de Interleucina-1/fisiología , Animales , Encéfalo/inmunología , Encéfalo/metabolismo , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Fatiga/metabolismo , Fatiga/patología , Inflamación/metabolismo , Inflamación/patología , Interleucina-1beta/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora , Transducción de Señal
20.
Oncogenesis ; 7(10): 81, 2018 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-30297705

RESUMEN

The incidence of human papillomavirus-associated head and neck squamous cell carcinoma (HPV[ + ] HNSCC) is rapidly increasing. Although clinical management of primary HPV( + ) HNSCC is relatively successful, disease progression, including recurrence and metastasis, is often fatal. Moreover, patients with progressive disease face limited treatment options and significant treatment-associated morbidity. These clinical data highlight the need to identify targetable mechanisms that drive disease progression in HPV( + ) HNSCC to prevent and/or treat progressive disease. Interestingly, ß-adrenergic signaling has recently been associated with pro-tumor processes in several disease types. Here we show that an aggressive murine model of recurrent/metastatic HPV( + ) HNSCC upregulates ß2-adrenergic receptor (ß2AR) expression, concordant with significantly heightened mitochondrial metabolism, as compared with the parental model from which it spontaneously derived. ß-Adrenergic blockade effectively inhibits in vitro proliferation and migratory capacity in this model, effects associated with an attenuation of hyperactive mitochondrial respiration. Importantly, propranolol, a clinically available nonselective ß-blocker, significantly slows primary tumor growth, inhibits metastatic development, and shows additive benefit alongside standard-of-care modalities in vivo. Further, via CRISPR/Cas9 technology, we show that the hyperactive mitochondrial metabolic profile and aggressive in vivo phenotype of this recurrent/metastatic model are dependent on ß2AR expression. These data implicate ß2AR as a modulator of mitochondrial metabolism and disease progression in HPV( + ) HNSCC, and warrant further investigation into the use of ß-blockers as low cost, relatively tolerable, complementary treatment options in the clinical management of this disease.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA