22 research outputs found

    Cannabinoids reduce ErbB2-driven breast cancer progression through Akt inhibition.

    Get PDF
    RIGHTS : This article is licensed under the BioMed Central licence at http://www.biomedcentral.com/about/license which is similar to the 'Creative Commons Attribution Licence'. In brief you may : copy, distribute, and display the work; make derivative works; or make commercial use of the work - under the following conditions: the original author must be given credit; for any reuse or distribution, it must be made clear to others what the license terms of this work are.BACKGROUND: ErbB2-positive breast cancer is characterized by highly aggressive phenotypes and reduced responsiveness to standard therapies. Although specific ErbB2-targeted therapies have been designed, only a small percentage of patients respond to these treatments and most of them eventually relapse. The existence of this population of particularly aggressive and non-responding or relapsing patients urges the search for novel therapies. The purpose of this study was to determine whether cannabinoids might constitute a new therapeutic tool for the treatment of ErbB2-positive breast tumors. We analyzed their antitumor potential in a well established and clinically relevant model of ErbB2-driven metastatic breast cancer: the MMTV-neu mouse. We also analyzed the expression of cannabinoid targets in a series of 87 human breast tumors. RESULTS: Our results show that both Delta9-tetrahydrocannabinol, the most abundant and potent cannabinoid in marijuana, and JWH-133, a non-psychotropic CB2 receptor-selective agonist, reduce tumor growth, tumor number, and the amount/severity of lung metastases in MMTV-neu mice. Histological analyses of the tumors revealed that cannabinoids inhibit cancer cell proliferation, induce cancer cell apoptosis, and impair tumor angiogenesis. Cannabinoid antitumoral action relies, at least partially, on the inhibition of the pro-tumorigenic Akt pathway. We also found that 91% of ErbB2-positive tumors express the non-psychotropic cannabinoid receptor CB2. CONCLUSIONS: Taken together, these results provide a strong preclinical evidence for the use of cannabinoid-based therapies for the management of ErbB2-positive breast cancer

    Cannabinoid receptor CB2 drives HER2 pro-oncogenic signaling in breast cancer

    Get PDF
    Pharmacological activation of cannabinoid receptors elicits antitumoral responses in different models of cancer. However, the biological role of these receptors in tumor physio-pathology is still unknown. We analyzed CB2 cannabinoid receptor protein expression in two series of 166 and 483 breast tumor samples operated in the University Hospitals of Kiel, Tübingen and Freiburg between 1997 and 2010. CB2 mRNA expression was also analyzed in previously published DNA microarray datasets. The role of CB2 in oncogenesis was studied by generating a mouse line that expresses the HER2 rat ortholog (neu) and lacks CB2, and by a variety of biochemical and cell biology approaches in human breast cancer cells in culture and in vivo, upon modulation of CB2 expression by si/shRNAs and overexpression plasmids. CB2-HER2 molecular interaction was studied by co-localization, coimmunoprecipitation and proximity ligation assays. We show an association between elevated CB2 expression in HER2+ breast tumors and poor patient prognosis. We also demonstrate that genetic inactivation of CB2 impairs tumor generation and progression in MMTV-neu mice. Moreover, we show that HER2 upregulates CB2 expression by activating the transcription factor ELK1 via the ERK cascade, and that an increased CB2 expression activates the HER2 prooncogenic signaling machinery at the level of the tyrosine kinase c-SRC. Finally, HER2 and CB2 form heteromers in cancer cells. Our findings reveal an unprecedented role of CB2 as a pivotal regulator of HER2 pro-oncogenic signaling in breast cancer, and suggest that CB2 may be a biomarker with prognostic value in these tumors

    Assessment of Cannabidiol and ∆9-Tetrahydrocannabiol in Mouse Models of Medulloblastoma and Ependymoma

    Get PDF
    Children with medulloblastoma and ependymoma are treated with a multidisciplinary approach that incorporates surgery, radiotherapy, and chemotherapy; however, overall survival rates for patients with high-risk disease remain unsatisfactory. Data indicate that plant-derived cannabinoids are effective against adult glioblastoma; however, preclinical evidence supporting their use in pediatric brain cancers is lacking. Here we investigated the potential role for ∆9- tetrahydrocannabinol (THC) and cannabidiol (CBD) in medulloblastoma and ependymoma. Dose- dependent cytotoxicity of medulloblastoma and ependymoma cells was induced by THC and CBD in vitro, and a synergistic reduction in viability was observed when both drugs were combined. Mechanistically, cannabinoids induced cell cycle arrest, in part by the production of reactive oxygen species, autophagy, and apoptosis; however, this did not translate to increased survival in orthotopic transplant models despite being well tolerated. We also tested the combination of cannabinoids with the medulloblastoma drug cyclophosphamide, and despite some in vitro synergism, no survival advantage was observed in vivo. Consequently, clinical benefit from the use of cannabinoids in the treatment of high-grade medulloblastoma and ependymoma is expected to be limited. This study emphasizes the importance of preclinical models in validating therapeutic agent efficacy prior to clinical trials, ensuring that enrolled patients are afforded the most promising therapies available

    Activation of the orphan receptor GPR55 by lysophosphatidylinositol promotes metastasis in triple-negative breast cancer

    Full text link
    The orphan G protein-coupled receptor GPR55 has been directly or indirectly related to basic alterations that drive malignant growth: uncontrolled cancer cell proliferation, sustained angiogenesis, and cancer cell adhesion and migration. However, little is known about the involvement of this receptor in metastasis. Here, we show that elevated GPR55 expression in human tumors is associated with the aggressive basal/triple-negative breast cancer population, higher probability to develop metastases, and therefore poor patient prognosis. Activation of GPR55 by its proposed endogenous ligand lysophosphatidylinositol confers pro-invasive features on breast cancer cells both in vitro and in vivo. Specifically, this effect is elicited by coupling to Gq/11 heterotrimeric proteins and the subsequent activation, through ERK, of the transcription factor ETV4/PEA3. Together, these data show that GPR55 promotes breast cancer metastasis, and supports the notion that this orphan receptor may constitute a new therapeutic target and potential biomarker in the highly aggressive triple-negative subtypeThis work was supported by grants from Spanish Ministry of Economy and Competitiveness [PI11/00295 to CS, PI14/01101 to CS and EP-G, SAF2013-46183-R to MQ, and SAF2014-54705-R to MV-M, supported with European Regional Development (FEDER) funds] and Madrid Regional Government (S2010/BMD-2308 to MG, and 2010/BMD-2359 to MQ). EPG was a recipient of a Postdoctoral Research Contract from Fundación Científica Asociación Española Contra el Cáncer and a Federation of the Societies of Biochemistry and Molecular Biology (FEBS) Short-term Fellowship. SB-B and SC-L are recipients of a Formación de Profesorado Universitario (FPU) fellowship and a Ramón y Cajal research contract, respectively, from the Spanish Ministry of Economy and Competitivenes

    Therapeutic targeting of HER2–CB2R heteromers in HER2-positive breast cancer

    Get PDF
    There is a subtype of breast cancer characterized by the overexpression of the oncogene HER2. Although most patients with this diagnosis benefit from HER2-targeted treatments, some do not respond to these therapies and others develop resistance with time. New tools are therefore warranted for the treatment of this patient population, and for early identification of those individuals at a higher risk of developing innate or acquired resistance to current treatments. Here, we show that HER2 forms heteromer complexes with the cannabinoid receptor CB2R, the expression of these structures correlates with poor patient prognosis, and their disruption promotes antitumor responses. Collectively, our results support HER2–CB2R heteromers as new therapeutic targets and prognostic tools in HER2+ breast cancer

    Papel del receptor huérfano GPR55 en la fisiopatología del cáncer : implicación en la proliferación e invasión de células tumorales

    Get PDF
    GPR55 es un receptor huérfano acoplado a proteinas G que, según publicaciones recientes, podría ser activado por lisofosfatidilinositol y determinados cannabinoides. Aunque se conoce poco todavía sobre su función fisiológica, existen evidencias que indican que podría desempeñar un papel importante en la regulación del metabolismo óseo o el control del dolor inflamatorio, por ejemplo. En esta Tesis hemos estudiado el papel de GPR55 en la fisiopatología del cáncer. Nuestros resultados muestran que GPR55 se expresa en tumores de distintos orígenes y que su expresión es significativamente más alta en tejidos transformados frente a tejidos sanos. Dicha expresión se correlaciona además con una mayor agresividad tumoral y un peor pronóstico de los pacientes. Los estudios que hemos realizado demuestran que GPR55 proporciona a las células cancerígenas una serie de ventajas adaptativas que podrían explicar la mayor agresividad de los tumores que expresan niveles elevados de GPR55. Primero, hemos demostrado que GPR55 induce la proliferación de células tumorales a través de la activación de la cascada de señalización ERK/MAPK, tanto en cultivos celulares como en modelos in vivo de cáncer.Esta activación promueve el crecimiento tumoral en modelos animales basados en xenografts y en el modelo de carcinogénesis química en piel de ratón. Segundo, hemos observado que la expresión de GPR55 favorece otra serie de características estrechamente relacionadas con la progresión tumoral en general y la generación de metástasis en particular. Así, GPR55 promueve el crecimiento independiente de anclaje, la migración e invasión de células tumorales a través de la activación de proteínas Gq y la generación in vivo de metástasis en pulmones. Por último, hemos investigado si GPR55 está implicado en el efecto bifásico de los cannabinoides sobre la proliferación de células tumorales. Nuestros resultados muestran que GPR55 participa en la respuesta pro-proliferativa que induce el THC a bajas concentraciones, y sugieren que dicha acción podría estar mediada por la interacción funcional entre GPR55 y el receptor clásico de cannabinoides CB1. En conjunto, estos resultados sugieren que GPR55 podría ser un nuevo marcador con valor pronóstico y una nueva diana terapéutica en oncología. GPR55 is an orphan G protein-coupled receptor that has been proposed to be engaged by lysophospholipids and cannabinoids. Though little is known about the physiological function of GPR55, emerging evidences point to important regulatory functions throughout the body. In this Thesis we studied the role of GPR55 in cancer physio-pathology. We found that GPR55 is expressed in human tumors from different origins, and that this expression is higher in the transformed tissues as compared with the corresponding non-transformed tissues and correlates with tumor aggressiveness. Our results show that GPR55 expression confers a series of adaptative advantages on cancer cells that make them more aggressive. First, we demonstrate that GPR55 drives cancer cell proliferation, through activation of the ERK/MAPK cascade, both in cell cultures and in vivo, which results in the promotion of tumor growth in xenograft-based and chemical-carcinogenesis animal models of cancer. Second, we show that GPR55 expression favors another set of capabilities that are intimately related to tumor progression in general and the generation of metastasis in particular. Thus, GPR55 promotes anchorage-independent growth, migration and invasion of cancer cells in culture through activation of Gq proteins and the generation of lung metastasis in vivo. Finally, we investigated if GPR55 is involved in the biphasic effect of cannabinoids on cancer cell proliferation (low concentrations producing pro-proliferative responses while high concentrations inducing anti-proliferative effects). Our data show that GPR55 modulates the proliferation-inducing response produced by low concentrations of cannabinoids, and suggest that this action may be produced by functional interaction of GPR55 with the classical cannabinoid receptor CB1. Together, our data suggests that GPR55 could be a new biomarker with prognostic value and a therapeutic target in oncology

    Activation of the orphan receptor GPR55 by lysophosphatidylinositol promotes metastasis in triple-negative breast cancer

    Get PDF
    et al.The orphan G protein-coupled receptor GPR55 has been directly or indirectly related to basic alterations that drive malignant growth: uncontrolled cancer cell proliferation, sustained angiogenesis, and cancer cell adhesion and migration. However, little is known about the involvement of this receptor in metastasis. Here, we show that elevated GPR55 expression in human tumors is associated with the aggressive basal/triple-negative breast cancer population, higher probability to develop metastases, and therefore poor patient prognosis. Activation of GPR55 by its proposed endogenous ligand lysophosphatidylinositol confers pro-invasive features on breast cancer cells both in vitro and in vivo. Specifically, this effect is elicited by coupling to G heterotrimeric proteins and the subsequent activation, through ERK, of the transcription factor ETV4/PEA3. Together, these data show that GPR55 promotes breast cancer metastasis, and supports the notion that this orphan receptor may constitute a new therapeutic target and potential biomarker in the highly aggressive triple-negative subtype.This work was supported by grants from Spanish Ministry of Economy and Competitiveness [PI11/00295 to CS, PI14/01101 to CS and EP-G, SAF2013-46183-R to MQ, and SAF2014-54705-R to MV-M, supported with European Regional Development (FEDER) funds] and Madrid Regional Government (S2010/BMD-2308 to MG, and 2010/ BMD-2359 to MQ). EPG was a recipient of a Postdoctoral Research Contract from Fundación Científica Asociación Española Contra el Cáncer and a Federation of the Societies of Biochemistry and Molecular Biology (FEBS) Short-term Fellowship. SB-B and SC-L are recipients of a Formación de Profesorado Universitario (FPU) fellowship and a Ramón y Cajal research contract, respectively, from the Spanish Ministry of Economy and Competitiveness.Peer Reviewe

    The Role of Cannabinoids as Anticancer Agents in Pediatric Oncology

    No full text
    Cannabinoids are a group of chemicals that bind to receptors in the human body and, in turn, modulate the endocannabinoid system (ECS). They can be endogenously produced, synthetic, or derived from the plant Cannabis sativa L. Research over the past several decades has shown that the ECS is a cellular communication network essential to maintain multiple biological functions and the homeostasis of the body. Indeed, cannabinoids have been shown to influence a wide variety of biological effects, including memory, pain, reproduction, bone remodeling or immunity, to name a few. Unsurprisingly, given these broad physiological effects, alterations of the ECS have been found in different diseases, including cancer. In recent years, the medical use of cannabis has been approved in different countries for a variety of human conditions. However, the use of these compounds, specifically as anticancer agents, remains controversial. Studies have shown that cannabinoids do have anticancer activity in different tumor types such as breast cancer, melanoma, lymphoma and adult brain cancer. Specifically, phytocannabinoids Δ9-tetrahydrocannabinol (THC) and cannabidiol (CBD) has been shown to induce apoptosis and inhibit proliferation of adult cancer cells, as well as modulate angiogenesis and metastasis. Despite increasing evidence that cannabinoids elicit antitumor effects in adult cancers, there is minimal data available on their effects in children or in pediatric cancers despite public and clinical demand for information. Here we describe a comprehensive and critical review of what is known about the effects of cannabinoids on pediatric cancers, highlight current gaps in knowledge and identify the critical issues that need addressing before considering these promising but controversial drugs for use in pediatric oncology

    The orphan receptor GPR55 drives skin carcinogenesis and is upregulated in human squamous cell carcinomas

    No full text
    G protein-coupled receptors (GPCRs) control crucial physiological processes and their dysfunction contributes to various human diseases, including cancer. The orphan GPCR GPR55 was identified and cloned more than a decade ago, but very little is known about its physio-pathological relevance. It has been recently shown that GPR55 controls the behavior of human cancer cell lines in culture and xenografts. However, the assessment of the actual role of this receptor in malignant transformation in vivo is hampered by the lack of studies on its functional impact in clinically-relevant models of cancer. Here we demonstrate that GPR55 drives mouse skin tumor development. Thus, GPR55-deficient mice were more resistant to DMBA/TPA-induced papilloma and carcinoma formation than their wild-type littermates. GPR55 exerted this pro-tumor effect primarily by conferring a proliferative advantage on cancer cells. In addition, GPR55 enhanced skin cancer cell anchorage-independent growth, invasiveness and tumorigenicity in vivo, suggesting that it promotes not only tumor development but also tumor aggressiveness. Finally, we observed that GPR55 is upregulated in human skin tumors and other human squamous cell carcinomas compared with the corresponding healthy tissues. Altogether, these findings reveal the pivotal importance of GPR55 in skin tumor development, and suggest that this receptor may be used as a new biomarker and therapeutic target in squamous cell carcinomas.This work was supported by grants from Fondo de Investigaciones Sanitarias (PI080253 to CS), Fundación Mutua Madrileña (to CS), GW Pharmaceuticals/Otsuka Pharmaceuticals (to CS), Comunidad de Madrid (S2010/BMD-2038 to MG) and Spanish Ministry of Science and Innovation (SAF2010-19152 to MQ and SAF2008-00121 and SAF2011-26122-C02- 01 to JMP). EPG and CA were the recipients of a Postdoctoral Research Contract from Fundación Científica Asociación Española Contra el Cáncer (AECC) and a PFIS PhD studentship from the Spanish Ministry of Science and Innovation, respectively.Peer Reviewe
    corecore