RUAN FELIPE VIEIRA MEDRANO

(Fonte: Lattes)
Índice h a partir de 2011
8
Projetos de Pesquisa
Unidades Organizacionais
LIM/24 - Laboratório de Oncologia Experimental, Hospital das Clínicas, Faculdade de Medicina

Resultados de Busca

Agora exibindo 1 - 10 de 17
  • conferenceObject
    Anti-tumor Effect of P19Arf and Interferon-beta Gene Transference to Mouse Melanoma and Mouse Lung Carcinoma Cells is Revealed by a Strong Bystander Activity and a Potential Immune Response
    (2012) RIBEIRO, A. H.; MEDRANO, R. F. V.; CATANI, J. P. P.; STRAUSS, B. E.
    Introduction: Two hallmarks of tumor progression are resistance to cell death and lack of an effective anti-tumor immune response. Loss of p53 function, by genetic mutation or alterations in its pathway, such as p19Arf loss and/or mdm2 over-expression, inhibits one of the primary coordinators of cell death. Interferon-beta (IFN), a stimulator of the immune response with anti-neoplastic functions, is also frequently lost in some tumor types. We propose that p19Arf and IFN gene transfer would be an effective strategy, especially in wild-type p53 tumor cells, since cell death and immune activation would be combined to combat the tumor at primary treatment site and metastasis. Material and Methods: Recombinant adenoviral vectors with RGD-modified fiber (rAdRGD) and a p53-responsive promoter (PG) were constructed containing genes of p19Arf, IFN or the combination of both. Evaluation of in vitro antiproliferative effect of transgenes in B16F10 cells (B16, mouse melanoma, p53 wt) was done by annexin/PI staining and MTT assays. Bystander effect was revealed by cell cycle analysis of populations transduced with different proportions of the viruses. Antitumor effect in vivo was observed by treatment of established LLC1 tumors (mouse lung carcinoma, p53 wt) with intratumoral injection of rAdRGD in C57BL/6 mice. Involvement of immune response was revealed by second tumor challenge at contralateral flank of mice with a developed and treated first tumor. Results and Discussion: Cell death was resulted from the p19Arf and IFN combined transference (74% subG0), yet single gene transfer yielded only half the number of subG0 cells. A similar result was seen by measurement of cell viability with MTT. Evidence on a bystander effect was revealed when approximately 50% subG0 cells were observed, even though only 10% of the cells had been transduced with IFN. In a population of cells transduced with p19Arf, when 10% of them also expressed IFN, the number of subG0 cells increased to 68%, compared to transduction of p19Arf alone, which results in 45% subG0 cells. This indicates that p19Arf can sensitize cells to death by IFN bystander effect. In vivo assays with the LLC1 model have shown that in situ gene therapy of p19Arf and IFN combination was more effective to inhibit tumor progression and increase survival than application of a single gene. These animals were then challenged with the implantation of a second tumor, revealing greater retardation of growth at the secondary tumor site in mice treated with the combined gene therapy at the primary tumor locus as compared to animals that received single gene treatment. Conclusion: The use of p53-responsive vectors to express p19Arf and IFN represents a potential strategy for melanoma and lung carcinoma tumor suppression. We have shown that complementation of the p53/Arf and interferon pathways in the primary tumor may generate a strong bystander effect as well as immune stimulation.
  • bookPart
    Imunologia de tumores
    (2015) MEDRANO, Ruan F. V.; RODRIGUES, Elaine Guadelupe
  • conferenceObject
    Use of p19Arf/interferon-beta immunotherapy in association with chemotherapy permits reduced drug dosage and avoids cardiotoxicity associated with doxorubicin
    (2019) STRAUSS, Bryan E.; MEDRANO, Ruan F. V.; TAMURA, Rodrigo; MENDONCA, Samir A.; FEITOSA, Valker A.; DARIOLLI, Rafael; SALLES, Thiago A.; HUNGER, Aline; CATANI, Joao P. P.; RODRIGUES, Elaine G.
  • conferenceObject
    Potentiation of doxorubicin low-dose efficacy through its association with p19Arf/Interferon-beta immunotherapy: Combining two immunogenic cell death inducers for the treatment of cancer.
    (2018) SR., Ruan F. V. Medrano; SR., Samir A. Mendonca; SR., Aline H. Ribeiro; SR., Joao P. P. Catani; SR., Valker A. Feitosa; SR., Elaine G. Rodrigues; SR., Bryan E. Strauss
  • article 21 Citação(ões) na Scopus
    Combined p19Arf and interferon-beta gene transfer enhances cell death of B16 melanoma in vitro and in vivo
    (2013) MERKEL, C. A.; MEDRANO, R. F. V.; BARAUNA, V. G.; STRAUSS, B. E.
    Approximately 90% of melanomas retain wild-type p53, a characteristic that may help shape the development of novel treatment strategies. Here, we employed an adenoviral vector where transgene expression is,controlled by p53 to deliver the p19 alternate reading frame (An) and interferon-beta (IFN beta) complementary DNAs in the B16 mouse model of melanoma. In vitro, cell death was enhanced by combined gene transfer (63.82 +/- 15.30% sub-GO cells); yet introduction of a single gene resulted in significantly fewer hypoploid cells (37.73 +/- 7.3% or 36.96 +/- 11.58%, p19Arf or IFN beta, respectively, P < 0.05). Annexin V staining and caspase-3 cleavage indicate a cell death mechanism consistent with apoptosis. Using reverse transcriptase quantitative PCR, we show that key transcriptional targets of p53 were upregulated in the presence of p19Arf, although treatment with IFN beta did not alter expression of the genes studied. In situ gene therapy revealed significant inhibition of subcutaneous tumors by IFN beta (571 +/- 25 mm(3)) or the combination of p19Arf and IFN beta (489 +/- 124 mm(3)) as compared with the LacZ control (1875 +/- 33 mm(3), P < 0.001); whereas p19Arf yielded an intermediate result (1053 +/- 169 mm(3), P < 0.01 vs control). However, only the combination was associated with increased cell death and prolonged survival (P < 0.01). As shown here, the combined transfer of p19Arf and IFN beta using p53-responsive vectors enhanced cell death both in vitro and in vivo.
  • article 8 Citação(ões) na Scopus
    AAVPG: A vigilant vector where transgene expression is induced by p53
    (2013) BAJGELMAN, Marcio C.; MEDRANO, Ruan F. V.; CARVALHO, Anna Carolina P. V.; STRAUSS, Bryan E.
    Using p53 to drive transgene expression from viral vectors may provide on demand expression in response to physiologic stress, such as hypoxia or DNA damage. Here we introduce AAVPG, an adeno-associated viral (AAV) vector where a p53-responsive promoter, termed PG, is used to control transgene expression. In vitro assays show that expression from the AAVPG-luc vector was induced specifically in the presence of functional p53 (1038 +/- 202 fold increase, p < 0.001). The AAVPG-Iuc vector was an effective biosensor of p53 activation in response to hypoxia (4.48 +/- 0.6 fold increase in the presence of 250 mu M CoCl2, p < 0.001) and biomechanical stress (253 +/- 0.4 fold increase with stretching, p < 0.05). In vivo, the vigilant nature of the AAVPG-luc vector was revealed after treatment of tumor-bearing mice with doxorubicin (pretreatment, 3.4 x 10(5) +/- 0.43 x 10(5) photons/s; post-treatment, 6.6 x 10(5) +/- 2.1 x 10(5) photons/s, p < 0.05). These results indicate that the AAVPG vector is an interesting option for detecting p53 activity both in vitro and in vivo.
  • article 122 Citação(ões) na Scopus
    Immunomodulatory and antitumor effects of type I interferons and their application in cancer therapy
    (2017) MEDRANO, Ruan F. V.; HUNGER, Aline; MENDONCA, Samir Andrade; BARBUTO, Jose Alexandre M.; STRAUSS, Bryan E.
    During the last decades, the pleiotropic antitumor functions exerted by type I interferons (IFNs) have become universally acknowledged, especially their role in mediating interactions between the tumor and the immune system. Indeed, type I IFNs are now appreciated as a critical component of dendritic cell (DC) driven T cell responses to cancer. Here we focus on IFN-alpha and IFN-beta, and their antitumor effects, impact on immune responses and their use as therapeutic agents. IFN-alpha/beta share many properties, including activation of the JAK-STAT signaling pathway and induction of a variety of cellular phenotypes. For example, type I IFNs drive not only the high maturation status of DCs, but also have a direct impact in cytotoxic T lymphocytes, NK cell activation, induction of tumor cell death and inhibition of angiogenesis. A variety of stimuli, including some standard cancer treatments, promote the expression of endogenous IFN-alpha/beta, which then participates as a fundamental component of immunogenic cell death. Systemic treatment with recombinant protein has been used for the treatment of melanoma. The induction of endogenous IFN-alpha/beta has been tested, including stimulation through pattern recognition receptors. Gene therapies involving IFN-alpha/beta have also been described. Thus, harnessing type I IFNs as an effective tool for cancer therapy continues to be studied.
  • conferenceObject
    Combined p19Arf and Interferon-beta Gene Therapy: Evidence of Immune Response in Murine Models of Melanoma and Lung Carcinoma
    (2013) STRAUSS, Bryan E.; MEDRANO, Ruan Felipe V.; RIBEIRO, Aline H.; CATANI, Joao Paulo P.; MERKEL, Christian A.
    Background: Our previous work has included the development of viral vectors where transgene expression is controlled by the transcriptional functions of the p53 tumor suppressor protein. Since wild-type p53 is frequently maintained in melanoma, we propose that such vectors may provide an opportunity for interplay between endogenous and exogenous factors. Transfer of p19Arf, a functional partner of p53, should help activate endogenous p53, thus supporting both vector expression and killing of tumor cells. Interferon-beta (IFNβ) is known to activate the immune system, induce apoptosis and inhibit angiogenesis. Moreover, interactions of the p53/Arf and IFN pathways have been reported. Previously we have shown that combined, but not individual, transfer of p19Arf and IFNβ mediated by our p53-responsive Ad5 vector induced massive cell death of B16 (mouse melanoma) both in vitro and in vivo. Objective: Our current aims include revealing involvement of the immune system in response to gene transfer protocols utilizing p53-responsive Ad5 or AdRGD vectors. Methods/Results: B16 cells with forced expression of CAR were transduced ex vivo with the Ad5 vectors (called AdPG) and implanted subcutaneously in C57BL/6 mice. Seven days later, these same mice received a challenge with fresh B16 cells implanted s.c. in the contralateral flank. Transfer of IFNβ alone or in combination with p19Arf reduced tumor formation at the sites of the vaccination and challenge. However, the combined treatment resulted in smaller tumors with delayed progression and prolonged survival. In parallel, RGD-modified adenoviral vectors, AdRGDPG, were constructed and shown to increase viral tropism as well as provide the expected synergy between p19Arf and IFNβ in CAR-negative B16 cells. This new set of AdRGDPG vectors was used in a model of in situ gene therapy of Lewis Lung Carcinoma (LLC) where tumors were first established s.c. in C57BL/6 mice then treated in vivo with six rounds of viral transduction. Treatment with IFNβ alone or in combination with p19Arf was effective in retarding tumor progression. Strikingly, s.c. challenge tumors implanted in the contralateral flank were inhibited especially well only in the animals previously treated with the combination of p19Arf and IFNβ. Alternatively, LLC cells were implanted s.c. in Balb/c nude mice and treated in situ. In this case, we did not observe a reduction in tumor progression in any of the conditions, indicating the importance of the adaptive immune system for tumor inhibition in response to our gene transfer strategy. Conclusion: In mouse tumor cell lines that retain wild-type p53, treatment with the combination of p19 Arf and IFNβ appears to involve the immune system, induce immunological memory and may provide an advantage over mono-gene therapy.
  • conferenceObject
    Combined transfer of p19Arf and interferon-beta genes to mouse melanoma cells causes LC3B-and caspase-3-independent cell death and alters the expression of critical genes
    (2015) RIBEIRO, Aline H.; VALLE, Paulo R. Del; MEDRANO, Ruan F. V.; FERRARI, Daniel G.; ZANATTA, Daniela B.; STRAUSS, Bryan E.
  • article 5 Citação(ões) na Scopus
    Perspectives for cancer immunotherapy mediated by p19Arf plus interferon-beta gene transfer
    (2018) STRAUSS, Bryan E.; SILVA, Gissele Rolemberg Oliveira; VIEIRA, Igor de Luna; CERQUEIRA, Otto Luiz Dutra; VALLE, Paulo Roberto Del; MEDRANO, Ruan Felipe Vieira; MENDONCA, Samir Andrade
    While cancer immunotherapy has gained much deserved attention in recent years, many areas regarding the optimization of such modalities remain unexplored, including the development of novel approaches and the strategic combination of therapies that target multiple aspects of the cancer-immunity cycle. Our own work involves the use of gene transfer technology to promote cell death and immune stimulation. Such immunogenic cell death, mediated by the combined transfer of the alternate reading frame (p14ARF in humans and p19Arf in mice) and the interferon-beta cDNA in our case, was shown to promote an antitumor immune response in mouse models of melanoma and lung carcinoma. With these encouraging results, we are now setting out on the road toward translational and preclinical development of our novel immunotherapeutic approach. Here, we outline the perspectives and challenges that we face, including the use of human tumor and immune cells to verify the response seen in mouse models and the incorporation of clinically relevant models, such as patient-derived xenografts and spontaneous tumors in animals. In addition, we seek to combine our immunotherapeutic approach with other treatments, such as chemotherapy or checkpoint blockade, with the goal of reducing dosage and increasing efficacy. The success of any translational research requires the cooperation of a multidisciplinary team of professionals involved in laboratory and clinical research, a relationship that is fostered at the Cancer Institute of Sao Paulo.