ROGER CHAMMAS

(Fonte: Lattes)
Índice h a partir de 2011
27
Projetos de Pesquisa
Unidades Organizacionais
Departamento de Radiologia, Faculdade de Medicina - Docente
LIM/05 - Laboratório de Poluição Atmosférica Experimental, Hospital das Clínicas, Faculdade de Medicina
LIM/24 - Laboratório de Oncologia Experimental, Hospital das Clínicas, Faculdade de Medicina - Líder

Resultados de Busca

Agora exibindo 1 - 10 de 44
  • article 12 Citação(ões) na Scopus
    Resistance Mechanisms Influencing Oncolytic Virotherapy, a Systematic Analysis
    (2021) BHATT, Darshak K.; CHAMMAS, Roger; DAEMEN, Toos
    Resistance to therapy is a frequently observed phenomenon in the treatment of cancer, and as with other cancer therapeutics, therapies based on oncolytic viruses also face the challenges of resistance, such as humoral and cellular antiviral responses, and tumor-associated interferon-mediated resistance. In order to identify additional mechanisms of resistance that may contribute to therapeutic failure, we developed a systematic search strategy for studies published in PubMed. We analyzed 6143 articles on oncolytic virotherapy and found that approximately 8% of these articles use resistance terms in the abstract and/or title. Of these 439 articles, 87 were original research. Most of the findings reported pertain to resistance mediated by tumor-cell-dependent interferon signaling. Yet, mechanisms such as epigenetic modifications, hypoxia-mediated inhibition, APOBEC-mediated resistance, virus entry barriers, and spatiotemporal restriction to viral spread, although not frequently assessed, were demonstrated to play a major role in resistance. Similarly, our results suggest that the stromal compartment consisting of, but not limited to, myeloid cells, fibroblasts, and epithelial cells requires more study in relation to therapy resistance using oncolytic viruses. Thus, our findings emphasize the need to assess the stromal compartment and to identify novel mechanisms that play an important role in conferring resistance to oncolytic virotherapy.
  • article 9 Citação(ões) na Scopus
    Exercise Training Preserves Myocardial Strain and Improves Exercise Tolerance in Doxorubicin-Induced Cardiotoxicity
    (2021) GOMES-SANTOS, Igor L.; JORDAO, Camila P.; PASSOS, Clevia S.; BRUM, Patricia C.; OLIVEIRA, Edilamar M.; CHAMMAS, Roger; CAMARGO, Anamaria A.; NEGRAO, Carlos E.
    Doxorubicin causes cardiotoxicity and exercise intolerance. Pre-conditioning exercise training seems to prevent doxorubicin-induced cardiac damage. However, the effectiveness of the cardioprotective effects of exercise training concomitantly with doxorubicin treatment remains largely unknown. To determine whether low-to-moderate intensity aerobic exercise training during doxorubicin treatment would prevent cardiotoxicity and exercise intolerance, we performed exercise training concomitantly with chronic doxorubicin treatment in mice. Ventricular structure and function were accessed by echocardiography, exercise tolerance by maximal exercise test, and cardiac biology by histological and molecular techniques. Doxorubicin-induced cardiotoxicity, evidenced by impaired ventricular function, cardiac atrophy, and fibrosis. Exercise training did not preserve left ventricular ejection fraction or reduced fibrosis. However, exercise training preserved myocardial circumferential strain alleviated cardiac atrophy and restored cardiomyocyte cross-sectional area. On the other hand, exercise training exacerbated doxorubicin-induced body wasting without affecting survival. Finally, exercise training blunted doxorubicin-induced exercise intolerance. Exercise training performed during doxorubicin-based chemotherapy can be a valuable approach to attenuate cardiotoxicity.
  • article 45 Citação(ões) na Scopus
    Emerging Autophagy Functions Shape the Tumor Microenvironment and Play a Role in Cancer Progression-Implications for Cancer Therapy
    (2020) BUSTOS, Silvina Odete; ANTUNES, Fernanda; RANGEL, Maria Cristina; CHAMMAS, Roger
    The tumor microenvironment (TME) is a complex environment where cancer cells reside and interact with different types of cells, secreted factors, and the extracellular matrix. Additionally, TME is shaped by several processes, such as autophagy. Autophagy has emerged as a conserved intracellular degradation pathway for clearance of damaged organelles or aberrant proteins. With its central role, autophagy maintains the cellular homeostasis and orchestrates stress responses, playing opposite roles in tumorigenesis. During tumor development, autophagy also mediates autophagy-independent functions associated with several hallmarks of cancer, and therefore exerting several effects on tumor suppression and/or tumor promotion mechanisms. Beyond the concept of degradation, new different forms of autophagy have been described as modulators of cancer progression, such as secretory autophagy enabling intercellular communication in the TME by cargo release. In this context, the synthesis of senescence-associated secretory proteins by autophagy lead to a senescent phenotype. Besides disturbing tumor treatment responses, autophagy also participates in innate and adaptive immune signaling. Furthermore, recent studies have indicated intricate crosstalk between autophagy and the epithelial-mesenchymal transition (EMT), by which cancer cells obtain an invasive phenotype and metastatic potential. Thus, autophagy in the cancer context is far broader and complex than just a cell energy sensing mechanism. In this scenario, we will discuss the key roles of autophagy in the TME and surrounding cells, contributing to cancer development and progression/EMT. Finally, the potential intervention in autophagy processes as a strategy for cancer therapy will be addressed.
  • article 7 Citação(ões) na Scopus
    Secretory Autophagy Forges a Therapy Resistant Microenvironment in Melanoma
    (2022) BUSTOS, Silvina Odete; SANTOS, Nathalia Leal; CHAMMAS, Roger; ANDRADE, Luciana Nogueira de Sousa
    Simple Summary Tumor microenvironment (TME) is a complex of many cell types and extracellular matrix that play an active role in regulating and sustaining melanoma tumor progression. In this context, the secretion of several molecules, by secretory autophagy or exosome release, stimulates the intercellular communication between the different components of the TME modulating tumor response. Here, we discuss the current awareness around the role of extracellular secretion in melanoma TME and also investigate the molecules related to these secretion pathways in melanoma progression using public databases. Melanoma is the most aggressive skin cancer characterized by high mutational burden and large heterogeneity. Cancer cells are surrounded by a complex environment, critical to tumor establishment and progression. Thus, tumor-associated stromal components can sustain tumor demands or impair cancer cell progression. One way to manage such processes is through the regulation of autophagy, both in stromal and tumor cells. Autophagy is a catabolic mechanism that provides nutrients and energy, and it eliminates damaged organelles by degradation and recycling of cellular elements. Besides this primary function, autophagy plays multiple roles in the tumor microenvironment capable of affecting cell fate. Evidence demonstrates the existence of novel branches in the autophagy system related to cytoplasmic constituent's secretion. Hence, autophagy-dependent secretion assembles a tangled network of signaling that potentially contributes to metabolism reprogramming, immune regulation, and tumor progression. Here, we summarize the current awareness regarding secretory autophagy and the intersection with exosome biogenesis and release in melanoma and their role in tumor resistance. In addition, we present and discuss data from public databases concerning autophagy and exosome-related genes as important mediators of melanoma behavior. Finally, we will present the main challenges in the field and strategies to translate most of the pre-clinical findings to clinical practice.
  • article 0 Citação(ões) na Scopus
    Exploring the Neandertal legacy of pancreatic ductal adenocarcinoma risk in Eurasians
    (2023) PICCARDI, Margherita; GENTILUOMO, Manuel; BERTONCINI, Stefania; PEZZILLI, Raffaele; EROSS, Balint; BUNDUC, Stefania; UZUNOGLU, Faik G.; TALAR-WOJNAROWSKA, Renata; VANAGAS, Tomas; SPERTI, Cosimo; OLIVERIUS, Martin; AOKI, Mateus Nobrega; ERMINI, Stefano; HUSSEIN, Tamas; BOGGI, Ugo; JAMROZIAK, Krzysztof; MAIELLO, Evaristo; MORELLI, Luca; VODICKOVA, Ludmila; FRANCO, Gregorio Di; LANDI, Stefano; SZENTESI, Andrea; LOVECEK, Martin; PUZZONO, Marta; TAVANO, Francesca; LAARHOVEN, Hanneke W. M. van; ZERBI, Alessandro; MOHELNIKOVA-DUCHONOVA, Beatrice; STOCKER, Hannah; COSTELLO, Eithne; CAPURSO, Gabriele; GINOCCHI, Laura; LAWLOR, Rita T.; VANELLA, Giuseppe; BAZZOCCHI, Francesca; IZBICKI, Jakob R.; LATIANO, Anna; BUENO-DE-MESQUITA, Bas; PISANI, Ruggero Ponz de Leon; SCHOETTKER, Ben; SOUCEK, Pavel; HEGYI, Peter; GAZOULI, Maria; HACKERT, Thilo; KUPCINSKAS, Juozas; POSKIENE, Lina; TACELLI, Matteo; ROTH, Susanne; CARRARA, Silvia; PERRI, Francesco; HLAVAC, Viktor; THEODOROPOULOS, George E.; BUSCH, Olivier R.; MAMBRINI, Andrea; EIJCK, Casper H. J. van; ARCIDIACONO, Paolo; SCARPA, Aldo; PASQUALI, Claudio; BASSO, Daniela; LUCCHESI, Maurizio; MILANETTO, Anna Caterina; NEOPTOLEMOS, John P.; CAVESTRO, Giulia Martina; JANCIAUSKAS, Dainius; CHEN, Xuechen; CHAMMAS, Roger; GOETZ, Mara; BRENNER, Hermann; ARCHIBUGI, Livia; DANNEMANN, Michael; CANZIAN, Federico; TOFANELLI, Sergio; CAMPA, Daniele
    BackgroundThe genomes of present-day non-Africans are composed of 1-3% of Neandertal-derived DNA as a consequence of admixture events between Neandertals and anatomically modern humans about 50-60 thousand years ago. Neandertal-introgressed single nucleotide polymorphisms (aSNPs) have been associated with modern human disease-related traits, which are risk factors for pancreatic ductal adenocarcinoma (PDAC), such as obesity, type 2 diabetes, and inflammation. In this study, we aimed at investigating the role of aSNPs in PDAC in three Eurasian populations.ResultsThe high-coverage Vindija Neandertal genome was used to select aSNPs in non-African populations from 1000 Genomes project phase 3 data. Then, the association between aSNPs and PDAC risk was tested independently in Europeans and East Asians, using existing GWAS data on more than 200 000 individuals. We did not find any significant associations between aSNPs and PDAC in samples of European descent, whereas, in East Asians, we observed that the Chr10p12.1-rs117585753-T allele (MAF = 10%) increased the risk to develop PDAC (OR = 1.35, 95%CI 1.19-1.54, P = 3.59 x 10(-6)), with a P-value close to a threshold that takes into account multiple testing.ConclusionsOur results show only a minimal contribution of Neandertal SNPs to PDAC risk.
  • article 2 Citação(ões) na Scopus
    Aerobic exercise training mitigates tumor growth and cancer-induced splenomegaly through modulation of non-platelet platelet factor 4 expression
    (2023) TOBIAS, Gabriel C.; GOMES, Joao L. P.; FERNANDES, Larissa G.; VOLTARELLI, Vanessa A.; ALMEIDA, Ney R. de; JANNIG, Paulo R.; SOUZA, Rodrigo W. Alves de; NEGRAO, Carlos E.; OLIVEIRA, Edilamar M.; CHAMMAS, Roger; ALVES, Christiano R. R.; BRUM, Patricia C.
    Exercise training reduces the incidence of several cancers, but the mechanisms underlying these effects are not fully understood. Exercise training can affect the spleen function, which controls the hematopoiesis and immune response. Analyzing different cancer models, we identified that 4T1, LLC, and CT26 tumor-bearing mice displayed enlarged spleen (splenomegaly), and exercise training reduced spleen mass toward control levels in two of these models (LLC and CT26). Exercise training also slowed tumor growth in melanoma B16F10, colon tumor 26 (CT26), and Lewis lung carcinoma (LLC) tumor-bearing mice, with minor effects in mammary carcinoma 4T1, MDA-MB-231, and MMTV-PyMT mice. In silico analyses using transcriptome profiles derived from these models revealed that platelet factor 4 (Pf4) is one of the main upregulated genes associated with splenomegaly during cancer progression. To understand whether exercise training would modulate the expression of these genes in the tumor and spleen, we investigated particularly the CT26 model, which displayed splenomegaly and had a clear response to the exercise training effects. RT-qPCR analysis confirmed that trained CT26 tumor-bearing mice had decreased Pf4 mRNA levels in both the tumor and spleen when compared to untrained CT26 tumor-bearing mice. Furthermore, exercise training specifically decreased Pf4 mRNA levels in the CT26 tumor cells. Aspirin treatment did not change tumor growth, splenomegaly, and tumor Pf4 mRNA levels, confirming that exercise decreased non-platelet Pf4 mRNA levels. Finally, tumor Pf4 mRNA levels are deregulated in The Cancer Genome Atlas Program (TCGA) samples and predict survival in multiple cancer types. This highlights the potential therapeutic value of exercise as a complementary approach to cancer treatment and underscores the importance of understanding the exercise-induced transcriptional changes in the spleen for the development of novel cancer therapies.
  • article 6 Citação(ões) na Scopus
    The PANcreatic Disease ReseArch (PANDoRA) consortium: Ten years' experience of association studies to understand the genetic architecture of pancreatic cancer
    (2023) CAMPA, Daniele; GENTILUOMO, Manuel; STEIN, Angelika; AOKI, Mateus Nobrega; OLIVERIUS, Martin; VODICKOVA, Ludmila; JAMROZIAK, Krzysztof; THEODOROPOULOS, George; PASQUALI, Claudio; GREENHALF, William; ARCIDIACONO, Paolo Giorgio; UZUNOGLU, Faik; PEZZILLI, Raffaele; LUCHINI, Claudio; PUZZONO, Marta; LOOS, Martin; GIACCHERINI, Matteo; KATZKE, Verena; MAMBRINI, Andrea; KIUDELIENE, Edita; FEDERICO, Kauffmann Emanuele; JOHANSEN, Julia; HUSSEIN, Tamas; MOHELNIKOVA-DUCHONOVA, Beatrice; EIJCK, Casper H. J. van; BRENNER, Hermann; FARINELLA, Riccardo; PEREZ, Juan Sainz; LOVECEK, Martin; BUECHLER, Markus W.; HLAVAC, Viktor; IZBICKI, Jakob R.; HACKERT, Thilo; CHAMMAS, Roger; ZERBI, Alessandro; LAWLOR, Rita; FELICI, Alessio; GOETZ, Mara; CAPURSO, Gabriele; GINOCCHI, Laura; GAZOULI, Maria; KUPCINSKAS, Juozas; CAVESTRO, Giulia Martina; VODICKA, Pavel; MOZ, Stefania; NEOPTOLEMOS, John P.; KUNOVSKY, Lumir; BOJESEN, Stig E.; CARRARA, Silvia; GIOFFREDA, Domenica; MORKUNAS, Egidijus; ABIAN, Olga; BUNDUC, Stefania; BASSO, Daniela; BOGGI, Ugo; WLODARCZYK, Barbara; SZENTESI, Andrea; VANELLA, Giuseppe; CHEN, Inna; BIJLSMA, Maarten F.; KIUDELIS, Vytautas; LANDI, Stefano; Ben Schoettker; CORRADI, Chiara; GIESE, Nathalia; KAAKS, Rudolf; PEDUZZI, Giulia; HEGYI, Peter; MORELLI, Luca; FURBETTA, Niccolo; SOUCEK, Pavel; LATIANO, Anna; TALAR-WOJNAROWSKA, Renata; LINDGAARD, Sidsel C.; DIJK, Frederik; MILANETTOJ, Anna Caterina; TAVANO, Francesca; CERVENA, Klara; EROSS, Balint; TESTONI, Sabrina G.; VERHAGEN-OLDENAMPSEN, Judith H. E.; MALECKA-WOJCIESKO, E.; COSTELLO, Eithne; SALVIA, Roberto; MAIELLO, Evaristo; ERMINI, Stefano; SPERTI, Cosimo; HOLLECZEK, Bernd; PERRI, Francesco; SKIECEVICIENE, Jurgita; ARCHIBUGI, Livia; LUCCHESI, Maurizio; RIZZATO, Cosmeri; CANZIAN, Federico
    Pancreatic cancer has an incidence that almost matches its mortality. Only a small number of risk factors and 33 susceptibility loci have been identified. so Moreover, the relative rarity of pancreatic cancer poses significant hurdles for research aimed at increasing our knowledge of the genetic mechanisms contributing to the disease. Additionally, the inability to adequately power research questions prevents small monocentric studies from being successful. Several consortia have been established to pursue a better understanding of the genetic architecture of pancreatic cancers. The Pancreatic disease research (PANDoRA) consortium is the largest in Europe. PANDoRA is spread across 12 European countries, Brazil and Japan, bringing together 29 basic and clinical research groups. In the last ten years, PANDoRA has contributed to the discovery of 25 susceptibility loci, a feat that will be instrumental in stratifying the population by risk and optimizing preventive strategies.
  • article 33 Citação(ões) na Scopus
    Platelet-activating factor (PAF) receptor as a promising target for cancer cell repopulation after radiotherapy
    (2017) SILVA JR., I. A. da; CHAMMAS, R.; LEPIQUE, A. P.; JANCAR, S.
    A major drawback of radiotherapy is the accelerated growth of the surviving tumor cells. Radiotherapy generates a variety of lipids that bind to the receptor for platelet-activating factor, expressed by cells in the tumor microenvironment. In the present study, using the TC-1 tumor cell line, we found that irradiation induced a twofold increase in receptor expression and generated agonists of receptor. Irradiated cells induced a 20-fold increase in live TC-1 proliferation in vitro. Furthermore, subcutaneous co-injection of irradiated TC-1 cells with TC-1 expressing luciferase (TC-1 fluc(+)) markedly increased TC-1 fluc(+) proliferation in a receptor-dependent way. Moreover we used a human carcinoma cell line not expressing the PAF receptor (KBM) and the same cell transfected with the receptor gene (KBP). Following co-injection of live KBP cells with irradiated KBM in RAG mice, the tumor growth was significantly increased compared with tumor formed following co-injection of live KBM with irradiated KBM. This tumor cell repopulation correlated with increased infiltration of tumor-promoting macrophages (CD206+). We propose that receptor represents a possible target for improving the efficacy of radiotherapy through inhibition of tumor repopulation.
  • article 24 Citação(ões) na Scopus
    Somatic mutations in early onset luminal breast cancer
    (2018) ENCINAS, G.; SABELNYKOVA, V. Y.; LYRA, E. C. de; KATAYAMA, M. L. H.; MAISTRO, S.; VALLE, P. W. M. V.; PEREIRA, G. F. L.; RODRIGUES, L. M.; SERIO, P. A. M. P.; GOUVêA, A. C. R. C. de; GEYER, F. C.; BASSO, R. A.; PASINI, F. S.; DIZ, M. P. E.; BRENTANI, M. M.; GóES, J. C. G. S.; CHAMMAS, R.; BOUTROS, P. C.; FOLGUEIRA, M. A. A. K.
    Breast cancer arising in very young patients may be biologically distinct; however, these tumors have been less well studied. We characterized a group of very young patients (≤ 35 years) for BRCA germline mutation and for somatic mutations in luminal (HER2 negative) breast cancer. Thirteen of 79 unselected very young patients were BRCA1/2 germline mutation carriers. Of the non-BRCA tumors, eight with luminal subtype (HER2 negative) were submitted for whole exome sequencing and integrated with 29 luminal samples from the COSMIC database or previous literature for analysis. We identified C to T single nucleotide variants (SNVs) as the most common base-change. A median of six candidate driver genes was mutated by SNVs in each sample and the most frequently mutated genes were PIK3CA, GATA3, TP53 and MAP2K4. Potential cancer drivers affected in the present non-BRCA tumors include GRHL2, PIK3AP1, CACNA1E, SEMA6D, SMURF2, RSBN1 and MTHFD2. Sixteen out of 37 luminal tumors (43%) harbored SNVs in DNA repair genes, such as ATR, BAP1, ERCC6, FANCD2, FANCL, MLH1, MUTYH, PALB2, POLD1, POLE, RAD9A, RAD51 and TP53, and 54% presented pathogenic mutations (frameshift or nonsense) in at least one gene involved in gene transcription. The differential biology of luminal early-age onset breast cancer needs a deeper genomic investigation. © Encinas et al.
  • article 0 Citação(ões) na Scopus
    Polymorphisms in transcription factor binding sites and enhancer regions and pancreatic ductal adenocarcinoma risk
    (2024) UENAL, Pelin; LU, Ye; BUENO-DE-MESQUITA, Bas; EIJCK, Casper H. J. van; TALAR-WOJNAROWSKA, Renata; SZENTESI, Andrea; GAZOULI, Maria; KREIVENAITE, Edita; TAVANO, Francesca; MALECKA-WOJCIESKO, Ewa; EROSS, Balint; OLIVERIUS, Martin; BUNDUC, Stefania; AOKI, Mateus Nobrega; VODICKOVA, Ludmila; BOGGI, Ugo; GIACCHERINI, Matteo; KONDRACKIENE, Jurate; CHAMMAS, Roger; PALMIERI, Orazio; THEODOROPOULOS, George E.; BIJLSMA, Maarten F.; BASSO, Daniela; MOHELNIKOVA-DUCHONOVA, Beatrice; SOUCEK, Pavel; IZBICKI, Jakob R.; KIUDELIS, Vytautas; VANELLA, Giuseppe; ARCIDIACONO, Paolo Giorgio; WLODARCZYK, Barbara; HACKERT, Thilo; SCHOETTKER, Ben; UZUNOGLU, Faik G.; BAMBI, Franco; GOETZ, Mara; HLAVAC, Viktor; BRENNER, Hermann; PERRI, Francesco; CARRARA, Silvia; LANDI, Stefano; HEGYI, Peter; DIJK, Frederike; MAIELLO, Evaristo; CAPRETTI, Giovanni; TESTONI, Sabrina Gloria Giulia; PETRONE, Maria Chiara; STOCKER, Hannah; ERMINI, Stefano; ARCHIBUGI, Livia; GENTILUOMO, Manuel; CAVESTRO, Giulia Martina; PEZZILLI, Raffaele; FRANCO, Gregorio Di; MILANETTO, Anna Caterina; SPERTI, Cosimo; NEOPTOLEMOS, John P.; MORELLI, Luca; VOKACOVA, Klara; PASQUALI, Claudio; LAWLOR, Rita T.; BAZZOCCHI, Francesca; KUPCINSKAS, Juozas; CAPURSO, Gabriele; CAMPA, Daniele; CANZIAN, Federico
    Genome-wide association studies (GWAS) are a powerful tool for detecting variants associated with complex traits and can help risk stratification and prevention strategies against pancreatic ductal adenocarcinoma (PDAC). However, the strict significance threshold commonly used makes it likely that many true risk loci are missed. Functional annotation of GWAS polymorphisms is a proven strategy to identify additional risk loci. We aimed to investigate single-nucleotide polymorphisms (SNP) in regulatory regions [transcription factor binding sites (TFBSs) and enhancers] that could change the expression profile of multiple genes they act upon and thereby modify PDAC risk. We analyzed a total of 12,636 PDAC cases and 43,443 controls from PanScan/PanC4 and the East Asian GWAS (discovery populations), and the PANDoRA consortium (replication population). We identified four associations that reached study-wide statistical significance in the overall meta-analysis: rs2472632(A) (enhancer variant, OR 1.10, 95%CI 1.06,1.13, p = 5.5 x 10-8), rs17358295(G) (enhancer variant, OR 1.16, 95%CI 1.10,1.22, p = 6.1 x 10-7), rs2232079(T) (TFBS variant, OR 0.88, 95%CI 0.83,0.93, p = 6.4 x 10-6) and rs10025845(A) (TFBS variant, OR 1.88, 95%CI 1.50,1.12, p = 1.32 x 10-5). The SNP with the most significant association, rs2472632, is located in an enhancer predicted to target the coiled-coil domain containing 34 oncogene. Our results provide new insights into genetic risk factors for PDAC by a focused analysis of polymorphisms in regulatory regions and demonstrating the usefulness of functional prioritization to identify loci associated with PDAC risk.