Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
1.
Cancers (Basel) ; 15(17)2023 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-37686576

RESUMO

Adverse effects are a common consequence of cytotoxic cancer treatments. Over the last two decades there have been significant advances in exploring the relationship between the gut microbiome and these adverse effects. Changes in the gut microbiome were shown in multiple clinical studies to be associated with the development of acute gastrointestinal adverse effects, including diarrhoea and mucositis. However, more recent studies showed that changes in the gut microbiome may also be associated with the long-term development of psychoneurological changes, cancer cachexia, and fatigue. Therefore, the aim of this review was to examine the literature to identify potential contributions and associations of the gut microbiome with the wide range of adverse effects from cytotoxic cancer treatments.

2.
Cancer Chemother Pharmacol ; 91(6): 507-521, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37162533

RESUMO

PURPOSE: Adverse effects following fluoropyrimidine-based chemotherapy regimens are common. However, there are no current accepted diagnostic markers for prediction prior to treatment, and the underlying mechanisms remain unclear. This study aimed to determine genetic and non-genetic predictors of adverse effects. METHODS: Genomic DNA was analyzed for 25 single nucleotide polymorphisms (SNPs). Demographics, comorbidities, cancer and fluoropyrimidine-based chemotherapy regimen types, and adverse effect data were obtained from clinical records for 155 Australian White participants. Associations were determined by bivariate analysis, logistic regression modeling and Bayesian network analysis. RESULTS: Twelve different adverse effects were observed in the participants, the most common severe adverse effect was diarrhea (12.9%). Bivariate analysis revealed associations between all adverse effects except neutropenia, between genetic and non-genetic predictors, and between 8 genetic and 12 non-genetic predictors with more than 1 adverse effect. Logistic regression modeling of adverse effects revealed a greater/sole role for six genetic predictors in overall gastrointestinal toxicity, nausea and/or vomiting, constipation, and neutropenia, and for nine non-genetic predictors in diarrhea, mucositis, neuropathy, generalized pain, hand-foot syndrome, skin toxicity, cardiotoxicity and fatigue. The Bayesian network analysis revealed less directly associated predictors (one genetic and six non-genetic) with adverse effects and confirmed associations between six adverse effects, eight genetic predictors and nine non-genetic predictors. CONCLUSION: This study is the first to link both genetic and non-genetic predictors with adverse effects following fluoropyrimidine-based chemotherapy. Collectively, we report a wealth of information that warrants further investigation to elucidate the clinical significance, especially associations with genetic predictors and adverse effects.


Assuntos
Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Neutropenia , Humanos , Fluoruracila , Teorema de Bayes , Austrália , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/etiologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/genética , Antimetabólitos , Neutropenia/induzido quimicamente , Neutropenia/epidemiologia , Diarreia/induzido quimicamente , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos
3.
Dev Med Child Neurol ; 65(8): 1029-1042, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-36740907

RESUMO

AIM: To identify and evaluate psychometric properties of assessment tools for assessing pain interference in children, adolescents, and adults with chronic pain and the inability to self-report. METHOD: The protocol was registered with PROSPERO (CRD42022310102). A search was run in MEDLINE, Embase, and PsycInfo (29th March 2022) to identify articles reporting psychometric properties of pain interference assessment tools for children, adolescents, and adults with chronic pain and the inability to objectively self-report pain. Retrieved studies were reviewed by two authors (MGS, LCF) and study quality was assessed using COSMIN. RESULTS: Psychometric properties of 10 pain interference tools were assessed from 33 studies. The Paediatric Pain Profile (PPP) had low-quality evidence for content validity and internal consistency with children and adolescents who are unable to self-report. No tools for adults had evidence for content validity and internal consistency. No tool had evidence for all nine psychometric properties. INTERPRETATION: The PPP is recommended for pain interference assessment in children and adolescents with chronic pain and the inability to self-report. Few tools are available for adults. Three tools for children (Patient-Reported Outcome Measurement Information System Pediatric Proxy Pain Interference Scale; Bath Adolescent Pain Questionnaire for Parents; modified Brief Pain Inventory-Proxy [mBPI]) and three tools for adults (Doloplus-2; Patient-Reported Outcome Measurement Information System Pain Interference Scale-proxy; Brief Pain Inventory-proxy) are promising but require further investigation.


Assuntos
Dor Crônica , Adolescente , Criança , Humanos , Adulto , Autorrelato , Dor Crônica/diagnóstico , Psicometria , Inquéritos e Questionários , Medição da Dor/métodos , Reprodutibilidade dos Testes
4.
J Control Release ; 343: 584-599, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35149142

RESUMO

Micro- and nano-scale particulate formulations are widely investigated towards improving the oral bioavailability of both biologics and drugs with low solubility and/or low intestinal permeability. Particulate formulations harnessing physiological intestinal transport pathways have recently yielded remarkably high oral bioavailabilities, illustrating the need for better understanding the specific pathways underpinning particle small intestinal absorption and the relative role of intestinal cells. Mechanistic knowledge has been hampered by the well acknowledged limitations of current in vitro, in vivo and ex vivo models relevant to the human intestinal physiology and the lack of standardization in studies reporting absorption data. Here we review the relevant literature and critically discusses absorption pathways with a focus on the role of specific intestinal epithelial and immune cells. We conclude that while Microfold (M) cells are a valid target for oral vaccines, enterocytes play a greater role in the systemic bioavailability of orally administrated particulate formulations, particularly within the sub-micron size range. We also comment on less-reported mechanisms such as paracellular permeability of particles, persorption due to cell damage and uptake by migratory immune cells.


Assuntos
Absorção Intestinal , Intestino Delgado , Administração Oral , Disponibilidade Biológica , Transporte Biológico , Humanos , Intestino Delgado/metabolismo , Permeabilidade
5.
Cancer Chemother Pharmacol ; 89(2): 275-281, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34854953

RESUMO

PURPOSE: Irinotecan can cause high levels of diarrhea caused by toxic injury to the gastrointestinal microenvironment. Toll-like receptor 4 (TLR4) and the gut microbiome have previously been implicated in gastrointestinal toxicity and diarrhea; however, the link between these two factors has not been definitively determined. We used a tumor-bearing, intestinal epithelial cell (IEC) TLR4 knockout model (Tlr4ΔIEC) to assess microbiome changes following irinotecan treatment. We then determined if a fecal microbiota transplant (FMT) between Tlr4ΔIEC and wild-type (WT) mice altered irinotecan-induced gastrointestinal toxicity. METHODS: MC-38 colorectal cancer cells were injected into WT and Tlr4ΔIEC mice. Fecal samples were collected prior to tumor inoculation, prior to irinotecan treatment and at cull. 16S rRNA gene sequencing was used to assess changes in the microbiome. Next, FMT was used to transfer the microbiome phenotype between Tlr4ΔIEC and WT mice prior to irinotecan treatment. Gastrointestinal toxicity symptoms were assessed. RESULTS: In study 1, there were no compositional differences in the microbiome between Tlr4ΔIEC and WT mice at baseline. However, predicted functional capacity of the microbiome was different between WT and Tlr4ΔIEC at baseline and post-irinotecan. In study 2, Tlr4ΔIEC mice were protected from grade 3 diarrhea. Additionally, WT mice who did not receive FMT had more colonic damage in the colon compared to controls (P = 0.013). This was not seen in Tlr4ΔIEC mice or WT mice who received FMT (P > 0.05). CONCLUSION: Tlr4ΔIEC and WT had no baseline compositional microbiome differences, but functional differences at baseline and following irinotecan. FMT altered some aspects of irinotecan-induced gastrointestinal toxicity.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Microbioma Gastrointestinal/efeitos dos fármacos , Irinotecano/farmacologia , Receptor 4 Toll-Like/genética , Animais , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Transplante de Microbiota Fecal/métodos , Gastroenteropatias/induzido quimicamente , Gastroenteropatias/microbiologia , Microbioma Gastrointestinal/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , RNA Ribossômico 16S , Inibidores da Topoisomerase I/farmacologia
6.
Integr Cancer Ther ; 19: 1534735420928493, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32493068

RESUMO

Small molecule receptor tyrosine kinase inhibitors (SM-TKIs) are among a group of targeted cancer therapies, intended to be more specific to cancer cells compared with treatments, such as chemotherapy, hence reducing adverse events. Unfortunately, many patients report high levels of diarrhea, the pathogenesis of which remains under investigation. In this article, we compare the current state of knowledge of the pathogenesis of chemotherapy-induced diarrhea (CID) in comparison to SM-TKI-induced diarrhea, and investigate how a similar research approach in both areas may be beneficial. To this end, we review evidence that both treatment modalities may interact with the gut microbiome, and as such the microbiome should be investigated for its ability to reduce the risk of diarrhea.


Assuntos
Antineoplásicos , Neoplasias Pulmonares , Microbiota , Antineoplásicos/efeitos adversos , Diarreia/induzido quimicamente , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Inibidores de Proteínas Quinases/efeitos adversos
7.
J Int Acad Periodontol ; 22(2): 18-27, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32224547

RESUMO

OBJECTIVES: This study investigated changes induced by Porphyromonas gingivalis and on gastrointestinal histology and gut microbiome in a mouse model of experimental periodontitis. The effect of probiotic Lactobacillus rhamnosus GG (LGG) in altering these changes was also investigated. METHODS: IThirty-six mice were allocated into six groups. Experimental alveolar bone loss was induced by oral inoculation with P. gingivalis and F. nucleatum. LGG was orally inoculated or orally gavaged. Gastrointestinal tissue changes were assessed using histological analysis and immunohistochemistry. Caecal microbiome was analysed by sequencing 16S rRNA genes of caecal content. RESULTS: Inoculation with P. gingivalis and F. nucleatum induced inflammation throughout gastrointestinal tract (p less than 0.05), increased expression of IL-6 in ileum (p = 0.052) and altered composition of caecal microbiome (p less than 0.05) in experimental mice compared to controls. Mice treated with LGG had reduced tissue inflammation in duodenum (p = 0.044) and lowered levels of IL-6 in ileum (p = 0.048) when compared with disease. LGG therapy influenced gut microbiome changes. CONCLUSION: P. gingivalis and F. nucleatum inoculation induced significant changes in intestinal inflammation and caecal microbiome. Oral gavage with LGG exerted a protective effect against intestinal inflammation and limited gut microbiome changes associated with P. gingivalis and F. nucleatum.


Assuntos
Microbioma Gastrointestinal , Lacticaseibacillus rhamnosus , Probióticos , Animais , Disbiose , Camundongos , RNA Ribossômico 16S
8.
Ann Gastroenterol ; 32(6): 584-592, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31700235

RESUMO

BACKGROUND: Ulcerative colitis (UC) is a lifelong inflammatory bowel disease characterized by periods of intense colonic inflammation leading to debilitating symptoms. Delivery methods of current UC treatments are suboptimal and associated with side effects. Silica particles are a potential alternative delivery method for UC therapeutics, given their promising drug-loading and safety profiles. However, it is unknown whether silica particles preferably accumulate at sites of colonic inflammation. This study aimed to correlate silica particle accumulation with colonic inflammation in a rat UC model. METHODS: Albino Wistar rats received 4.5% dextran sulfate sodium (DSS) in drinking water (n=6) for 7 days to induce UC. Control rats (n=6) received drinking water only. UC activity was assessed daily using disease activity index. All rats were orally gavaged with silica particles labeled with Alexa-633 tags on day 9, followed by imaging at 3, 6, and 24 h. Silica particle distribution and accumulation were examined using biophotonic imaging, confocal microscopy and fluorescent spectrophotometry. Rats were killed on day 10, with jejunum, ileum and colon collected for histopathological scoring and quantification of fluorescence. RESULTS: Rats treated with DSS had significantly higher UC disease activity (P=0.033) and colonic histopathological scores (P=0.0087) compared to controls. No statistically significant between-group differences in silica particle accumulation were seen on live imaging or tissue analysis. CONCLUSIONS: No correlation was seen between silica particle accumulation and colonic inflammation. However to draw clear conclusions, further research is required to establish the potential of silica particles as a UC-targeted delivery method.

9.
Support Care Cancer ; 27(10): 4011-4022, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31286233

RESUMO

PURPOSE: The aim of this study was to update the clinical practice guidelines for the use of agents for the prevention and/or treatment of gastrointestinal mucositis (GIM). METHODS: A systematic review was conducted by the Mucositis Study Group of the Multinational Association of Supportive Care in Cancer/International Society for Oral Oncology (MASCC/ISOO). The body of evidence for each intervention, in each cancer treatment setting, was assigned an evidence level. Based on the evidence level, one of the following three guideline determinations was possible: Recommendation, Suggestion, and No Guideline Possible. RESULTS: A total of 78 papers across 13 interventions were examined of which 25 were included in the final review. No new guidelines were possible for any agent due to inadequate and/or conflicting evidence. Existing guidelines for probiotics and hyperbaric oxygen were unchanged. CONCLUSIONS: Of the agents studied for the prevention and treatment of GIM, the evidence continues to support use of probiotics containing Lactobacillus spp. for prevention of chemoradiotherapy and radiotherapy-induced diarrhea in patients with pelvic malignancy, and hyperbaric oxygen therapy to treat radiation-induced proctitis. Additional well-designed research is encouraged to enable a decision regarding palifermin, glutamine, sodium butyrate, and dietary interventions, for the prevention or treatment of GIM.


Assuntos
Quimiorradioterapia/efeitos adversos , Mucosite/tratamento farmacológico , Mucosite/prevenção & controle , Guias de Prática Clínica como Assunto , Proctite/tratamento farmacológico , Estomatite/tratamento farmacológico , Ácido Butírico/uso terapêutico , Fator 7 de Crescimento de Fibroblastos/uso terapêutico , Glutamina/uso terapêutico , Humanos , Oxigenoterapia Hiperbárica , Neoplasias/tratamento farmacológico
11.
Exp Biol Med (Maywood) ; 244(7): 554-564, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30907132

RESUMO

IMPACT STATEMENT: The study of M cells, a specialized epithelial cell type found in the follicle-associated epithelium, is hampered by the lack of a universal M cell marker. As such, many studies lack reliable and universally recognized methods to identify M cells in their proposed models. As a result of this it is difficult to ascertain whether the effects observed are due to the presence of M cells or an unaccounted variable. The outcome of this review is the thorough evaluation of the many M cell markers that have been used in the literature thus far and a proposed criterion for the identification of M cells for future publications. This will hopefully lead to an improvement in the quality of future publications in this field.


Assuntos
Células Epiteliais/metabolismo , Mucosa Intestinal/citologia , Tecido Linfoide/citologia , Animais , Separação Celular/métodos , Células Epiteliais/classificação , Células Epiteliais/citologia , Humanos , Mucosa Intestinal/metabolismo , Tecido Linfoide/metabolismo , Cultura Primária de Células/métodos , Proteoma/genética , Proteoma/metabolismo , Roedores , Especificidade da Espécie
12.
Int J Cancer ; 144(10): 2365-2376, 2019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-30155890

RESUMO

Chemotherapy-induced gastrointestinal toxicity (CIGT) occurs in up to 80% of all patients undergoing cancer treatment, and leads to symptoms such as diarrhoea, abdominal bleeding and pain. There is currently limited understanding of how to predict an individual patient's risk of CIGT. It is believed the gut microbiome and its interactions with the host's innate immune system plays a key role in the development of this toxicity and potentially other toxicities, however comprehensive bioinformatics modelling has not been rigorously performed. The innate immune system is strongly influenced by the microbial environment and vice-versa. Ways this may occur include the immune system controlling composition and compartmentalisation of the microbiome, the microbiome affecting development of antigen-presenting cells, and finally, the NLRP6 inflammasome orchestrating the colonic host-microbiome interface. This evidence calls into question the role of pre-treatment risk factors in the development of gastrointestinal toxicity after chemotherapy. This review aims to examine evidence of a bidirectional interaction between the gut microbiome and innate immunity, and how these interactions occur in CIGT. In the future, knowledge of these interactions may lead to improved personalised cancer medicine, predictive risk stratification methods and the development of targeted interventions to reduce, or even prevent, CIGT severity.


Assuntos
Antineoplásicos/efeitos adversos , Gastroenteropatias/induzido quimicamente , Gastroenteropatias/imunologia , Microbioma Gastrointestinal/imunologia , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Imunidade Inata/imunologia , Animais , Células Apresentadoras de Antígenos/imunologia , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Gastroenteropatias/microbiologia , Humanos , Sistema Imunitário/imunologia , Microbiota/imunologia , Neoplasias/tratamento farmacológico , Neoplasias/imunologia
13.
Cancer Chemother Pharmacol ; 83(2): 217-236, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30474704

RESUMO

PURPOSE: Severe gastrointestinal (GI) toxicity is a common adverse effect following 5-fluorouracil (5-FU)-based chemotherapy treatment. The presence of severe GI toxicity leads to treatment revisions, sub-optimal therapy outcomes, and decreases to patients' quality of life. There are no adequate predictors for 5-FU-induced severe GI toxicity risk. The Toll-like receptor/interleukin-1 (TIR) domain innate immune signalling pathway is known to be a mediating pathway in the development of GI toxicity. Hence, genetic variability in this signalling pathway may alter the pathophysiology of GI toxicity and, therefore, be predictive of risk. However, little research has investigated the effects of TIR domain innate immune signalling pathway single nucleotide polymorphism (SNPs) on the risk and development of severe GI toxicity. METHODS: This critical review surveyed the literature and reported on the in vitro, ex vivo and in vivo effects, as well as the genetic association, of selected TIR domain innate immune signalling pathway SNPs on disease susceptibility and gene functioning. RESULTS: Of the TIR domain innate immune signalling pathway SNPs reviewed, evidence suggests interleukin-1 beta (IL1B) and tumour necrosis factor alpha (TNF) SNPs have the greatest potential as predictors for severe GI toxicity risk. These results warrant further research into the effect of IL1B and TNF SNPs on the risk and development of severe GI toxicity. CONCLUSIONS: SNPs of the TIR domain innate immune signalling pathway have profound effects on disease susceptibility and gene functioning, making them candidate predictors for severe GI toxicity risk. The identification of a predictor for 5-FU-induced severe GI toxicity will allow the personalization of supportive care measures.


Assuntos
Fluoruracila/efeitos adversos , Gastroenteropatias/diagnóstico , Interleucina-1/genética , Neoplasias/tratamento farmacológico , Polimorfismo de Nucleotídeo Único , Receptores Toll-Like/genética , Fator de Necrose Tumoral alfa/genética , Antimetabólitos Antineoplásicos/efeitos adversos , Gastroenteropatias/induzido quimicamente , Gastroenteropatias/genética , Marcadores Genéticos , Humanos , Imunidade Inata , Valor Preditivo dos Testes , Domínios Proteicos
14.
Int J Radiat Biol ; 94(7): 645-655, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29855218

RESUMO

PURPOSE: Radiotherapy-induced gut toxicity (RIGT) is a debilitating effect of radiotherapy for cancer, often resulting in significant diarrhea and pain. Previous studies have highlighted roles of the intestinal microvasculature and matrix metalloproteinases (MMPs) in the development of RIGT. We hypothesized vascular mediators would be significantly altered in a dark agouti (DA) rat model of RIGT. Additionally, we aimed to assess the effect of MMP-2 and -9 inhibition on the response of tumor-associated microvascular endothelial cells (TAMECs) to radiation. METHODS: DA rats were administered 2.5 Gy abdominal irradiation (3 times/week over 6 weeks). Vascular endothelial growth factor (VEGF), transforming growth factor beta (TGFß), von Willebrand factor (VWF), angiostatin, and endostatin expression was assessed at 3, 6, and 15 weeks. Additionally, DA rat mammary adenocarcinoma tumor-associated microvascular endothelial cells (TAMECs) were used to assess the effects of radiation (12 Gy) and the MMP inhibitor SB-3CT on MMP, VEGF, and TGFß expression, and cell viability. RESULTS: VEGF mRNA expression was significantly increased in the colon at week 15 (p = .0012), and TGFß mRNA expression was significantly increased in both the jejunum and colon at week 3 (p = .0280 and p = .0310, respectively). Endostatin immunostaining was significantly increased at week 3 (p = .0046), and angiostatin at 3 and 6 weeks (p = .0022 and p = .0135, respectively). MMP-2 and -9 mRNA and total protein levels were significantly increased following irradiation of TAMECs. Although this increase was significantly attenuated by SB-3CT, it did not significantly alter endothelial cell viability or VEGF and TGFß mRNA expression. CONCLUSIONS: Findings of this study support the involvement of VEGF, TGFß, angiostatin, endostatin, and MMP-2 in the pathobiology of RIGT. However, the relationship between these mediators is complex and needs further investigation to improve understanding of their therapeutic potential in RIGT.


Assuntos
Trato Gastrointestinal/efeitos da radiação , Radioterapia/efeitos adversos , Angiostatinas/análise , Angiostatinas/fisiologia , Animais , Fracionamento da Dose de Radiação , Endostatinas/análise , Endostatinas/fisiologia , Feminino , Trato Gastrointestinal/química , Compostos Heterocíclicos com 1 Anel/farmacologia , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 9 da Matriz/genética , Ratos , Sulfonas/farmacologia , Fator de Crescimento Transformador beta/análise , Fator de Crescimento Transformador beta/fisiologia , Fator A de Crescimento do Endotélio Vascular/análise , Fator A de Crescimento do Endotélio Vascular/fisiologia
15.
Support Care Cancer ; 26(11): 3873-3882, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29754212

RESUMO

PURPOSE: Radiotherapy-induced gut toxicity (RIGT) is associated with significant diarrhoea, pain and rectal bleeding. Matrix metalloproteinases (MMPs) have been reported to be involved in chemotherapy-induced gut toxicity and RIGT following single-dose irradiation in vivo. We therefore proposed MMPs would be involved in the pathobiology of RIGT following fractionated irradiation. METHODS: Dark Agouti rats were treated with fractionated radiation (3 × 2.5 Gy/week for 6 weeks). Rats were killed at 3, 6 and 15 weeks to represent acute and chronic toxicities. Sections of jejunum and colon were immunostained for MMP-1, MMP-2, MMP-9 and MMP-14. Relative mRNA expression in jejunum and colon was quantified by RT-PCR for MMP-1, MMP-2, MMP-9 and MMP-14. Western blotting was also conducted on jejunum and colon tissue collected at week 6 to determine protein levels of pro- and active MMP-2. RESULTS: MMP-2 total protein levels, determined by western blotting, significantly increased in both the jejunum (p = 0.0359) and the colon (p = 0.0134) 6 weeks into the fractionated radiation schedule. MMP-1, MMP-2, and MMP-14 mRNA expression significantly increased in the jejunum. MMP-2 mRNA expression was also significantly increased in the colon. Immunostaining of MMP-2 was observed to be increased in both crypt enterocytes and the lamina propria. CONCLUSIONS: MMP-2 plays a role in the pathobiology of gastrointestinal toxicities following fractionated irradiation. Whilst MMP-1 and MMP-14 mRNA expression was increased, this occurred only in the jejunum, suggesting MMPs are differentially involved in RIGT depending on the intestinal region. Further studies are needed to elucidate the role these mediators play in the development and potentiation of RIGT.


Assuntos
Intestino Grosso/metabolismo , Intestino Grosso/efeitos da radiação , Intestino Delgado/metabolismo , Intestino Delgado/efeitos da radiação , Metaloproteinases da Matriz/genética , Lesões por Radiação/genética , Animais , Fracionamento da Dose de Radiação , Relação Dose-Resposta à Radiação , Feminino , Gastroenteropatias/etiologia , Gastroenteropatias/genética , Regulação Enzimológica da Expressão Gênica/efeitos da radiação , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Mucosa Intestinal/efeitos da radiação , Intestino Grosso/patologia , Intestino Delgado/patologia , Metaloproteinases da Matriz/metabolismo , Doses de Radiação , Lesões por Radiação/patologia , Ratos , Ratos Transgênicos
16.
Chemotherapy ; 63(5): 284-292, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30731451

RESUMO

BACKGROUND: The common cytotoxic mechanisms that underpin chemoefficacy and toxicity have hampered efforts to deliver effective supportive care interventions, particularly for gastrointestinal (GI) toxicity. Matrix metalloproteinases (MMPs) have been implicated in both tumor growth and GI toxicity, and as such MMP inhibitors present as a novel therapeutic avenue to simultaneously enhance treatment efficacy and reduce toxicity. OBJECTIVES: The aim of this study was to determine the efficacy of an MMP-9/12 inhibitor, AZD3342, on tumor growth and GI toxicity in a rat model. METHODS: Female tumor-bearing Dark Agouti rats (n = 90) were divided into 4 groups: vehicle control; methotrexate (MTX); AZD3342, and MTX + AZD3342. Tumors were measured daily (for 5 days) using digital calipers. GI toxicity was assessed using well-established clinical markers (diarrhea/weight loss), histopathological analysis, and functional assessment of intestinal barrier permeability. RESULTS: AZD3342 delayed the onset of severe diarrhea by 1 day (vs. MTX) but was unable to improve the overall severity of diarrhea. No changes were detected in tissue morphology or intestinal barrier function. AZD3342 alone suppressed tumor growth (p = 0.003 vs. vehicle) but did not enhance the efficacy of MTX. CONCLUSIONS: This study showed partial efficacy of AZD3342 in reducing tumor growth and delaying the onset of severe diarrhea caused by MTX in rats. We suggest further studies be undertaken targeting appropriate scheduling of AZD3342 as well as investigating different cytotoxic therapies that strongly activate MMP signaling.


Assuntos
Inibidores de Metaloproteinases de Matriz/uso terapêutico , Neoplasias/tratamento farmacológico , Compostos Orgânicos/uso terapêutico , Animais , Antimetabólitos Antineoplásicos/efeitos adversos , Antimetabólitos Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Diarreia/etiologia , Feminino , Humanos , Mucosa Intestinal/patologia , Inibidores de Metaloproteinases de Matriz/efeitos adversos , Metaloproteinases da Matriz/química , Metaloproteinases da Matriz/metabolismo , Metotrexato/efeitos adversos , Metotrexato/uso terapêutico , Neoplasias/patologia , Compostos Orgânicos/efeitos adversos , Distribuição Aleatória , Ratos , Transplante Heterólogo
17.
Int J Cancer ; 142(2): 369-380, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-28921512

RESUMO

Dacomitinib, an irreversible small-molecule pan-ErbB TKI, has a high incidence of diarrhea, which has been suggested to be due to chloride secretory mechanisms. Based on this hypothesis, crofelemer, an antisecretory agent may be an effective intervention. T84 monolayers were treated with 1 µM dacomitinib and 10 µM crofelemer, and mounted into Ussing chambers for electrogenic ion analysis. Crofelemer attenuated increases in chloride secretion in cells treated with dacomitinib. Albino Wistar rats (n = 48) were treated with 7.5 mg/kg dacomitinib and/or 25 mg/kg crofelemer via oral gavage for 21 days. Crofelemer significantly worsened dacomitinib-induced diarrhea (p = 0.0003), and did not attenuate weight loss (p < 0.0001). Sections of the ileum and colon were mounted into Ussing chambers, and secretory processes analyzed. This indicated that crofelemer lost its anti-secretory action in the presence of dacomitinib in this model. Mass spectrometry revealed that crofelemer did not change serum concentration of dacomitinib. Serum FITC dextran levels indicated that crofelemer was unable to attenuate dacomitinib-induced barrier dysfunction. Tight junction proteins were visualized with immunofluorescence. Qualitative analysis showed dacomitinib induced proteolysis of ZO-1 and occludin, and internalization of claudin-1, which was not attenuated by crofelemer. Detailed histopathological analysis showed that crofelemer was unable to attenuate dacomitinib-induced ileal damage. Crofelemer worsened dacomitinib-induced diarrhea, suggesting that antisecretory drug therapy may be ineffective in this setting.


Assuntos
Cloretos/metabolismo , Diarreia/tratamento farmacológico , Proantocianidinas/farmacologia , Quinazolinonas/toxicidade , Animais , Permeabilidade da Membrana Celular/efeitos dos fármacos , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Diarreia/induzido quimicamente , Diarreia/metabolismo , Eletrofisiologia , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/patologia , Humanos , Masculino , Ratos , Ratos Wistar , Células Tumorais Cultivadas , Redução de Peso/efeitos dos fármacos
18.
J Clin Periodontol ; 45(2): 204-212, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29121411

RESUMO

AIM: This study investigated the role of Lactobacillus rhamnosus GG (LGG) on bone loss and local and systemic inflammation in an in vivo mouse model of experimental periodontitis (PD). MATERIALS AND METHODS: Experimental PD was induced in mice by oral inoculation with Porphyromonas gingivalis and Fusobacterium nucleatum over a period of 44 days. The probiotic LGG was administered via oral inoculation or oral gavage prior to, and during disease induction. The antimicrobial activity of LGG on the inoculum was also tested. Alveolar bone levels and gingival tissue changes were assessed using in vivo microcomputed tomography and histological analysis. Serum levels of mouse homologues for IL-8 were measured using multiplex assays. RESULTS: Pre-treatment with probiotics either via oral gavage or via oral inoculation significantly reduced bone loss (p < .0001) and gingival inflammation (p < .0001) when compared with PD group. Oral gavage treatment group had significantly less tartrate-resistant acid phosphatase positive cells (p < .02) then PD group. LGG showed no antimicrobial activity against P. gingivalis and F. nucleatum. CONCLUSIONS: Lactobacillus rhamnosus GG effectively suppresses bone loss in a mouse model of induced PD irrespective of the mode of administration.


Assuntos
Perda do Osso Alveolar/prevenção & controle , Lacticaseibacillus rhamnosus , Periodontite/prevenção & controle , Probióticos/uso terapêutico , Animais , Modelos Animais de Doenças , Feminino , Fusobacterium nucleatum , Camundongos , Camundongos Endogâmicos BALB C , Periodontite/microbiologia , Porphyromonas gingivalis , Probióticos/administração & dosagem
19.
Int J Cancer ; 140(12): 2820-2829, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28316082

RESUMO

Dacomitinib-an irreversible pan-ErbB tyrosine kinase inhibitor (TKI)-causes diarrhoea in 75% of patients. Dacomitinib-induced diarrhoea has not previously been investigated and the mechanisms remain poorly understood. The present study aimed to develop an in-vitro and in-vivo model of dacomitinib-induced diarrhoea to investigate underlying mechanisms. T84 cells were treated with 1-4 µM dacomitinib and resistance and viability were measured using transepithelial electrical resistance (TEER) and XTT assays. Rats were treated with 7.5 mg/kg dacomitinib daily via oral gavage for 7 or 21 days (n = 6/group). Weights, and diarrhoea incidence were recorded daily. Rats were administered FITC-dextran 2 hr before cull, and serum levels of FITC-dextran were measured and serum biochemistry analysis was conducted. Detailed histopathological analysis was conducted throughout the gastrointestinal tract. Gastrointestinal expression of ErbB1, ErbB2 and ErbB4 was analysed using RT-PCR. The ileum and the colon were analysed using multiplex for expression of various cytokines. T84 cells treated with dacomitinib showed no alteration in TEER or cell viability. Rats treated with dacomitinib developed severe diarrhoea, and had significantly lower weight gain. Further, dacomitinib treatment led to severe histopathological injury localised to the ileum. This damage coincided with increased levels of MCP1 in the ileum, and preferential expression of ErbB1 in this region compared to all other regions. This study showed dacomitinib induces severe ileal damage accompanied by increased MCP1 expression, and gastrointestinal permeability in rats. The histological changes were most pronounced in the ileum, which was also the region with the highest relative expression of ErbB1.


Assuntos
Diarreia/induzido quimicamente , Trato Gastrointestinal/efeitos dos fármacos , Íleo/efeitos dos fármacos , Quinazolinonas/toxicidade , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Quimiocina CCL2/metabolismo , Neoplasias Colorretais/patologia , Diarreia/fisiopatologia , Modelos Animais de Doenças , Receptores ErbB/genética , Receptores ErbB/metabolismo , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/fisiopatologia , Expressão Gênica/efeitos dos fármacos , Humanos , Íleo/metabolismo , Íleo/fisiopatologia , Imuno-Histoquímica , Masculino , Permeabilidade/efeitos dos fármacos , Quinazolinonas/farmacologia , Ensaio de Radioimunoprecipitação , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
Curr Opin Support Palliat Care ; 11(2): 133-137, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28333868

RESUMO

PURPOSE OF REVIEW: Gastrointestinal toxicities are commonly reported following treatment with proteasome inhibitors. The first-generation proteasome inhibitor, bortezomib, induces significant gastrointestinal side effects including nausea, vomiting, diarrhoea, and constipation, occurring in up to 84% of patients. Despite the development of safer proteasome inhibitors, such as carfilzomib, gastrointestinal toxicities remain some of the most common side effects. This review aims to summarize the previous literature on proteasome inhibitor-induced gastrointestinal toxicities, report on recent updates in the field, and investigate possible mechanisms of this toxicity. RECENT FINDINGS: Updates in the literature have included a direct comparison of the safety of approved proteasome inhibitors, bortezomib and carfilzomib, reporting less neurotoxicity and similar gastrointestinal toxicity, from carfilzomib when compared with bortezomib. Many recent studies have investigated the safety of orally bioavailable proteasome inhibitors, such as ixazomib and oprozomib. However, little progress has been made in understanding the possible mechanisms of proteasome inhibitor-induced gastrointestinal toxicities. SUMMARY: Although recent studies have continued to report gastrointestinal toxicities resulting from proteasome inhibitor treatment, particularly when combined with other agents or when administered orally, the mechanisms of proteasome inhibitor-induced gut toxicity remain largely unexplored. Further studies are needed to investigate the pathophysiology of this toxicity to improve the safety of existing and novel proteasome inhibitors.


Assuntos
Antineoplásicos/efeitos adversos , Gastroenteropatias/induzido quimicamente , Inibidores de Proteassoma/efeitos adversos , Antineoplásicos/farmacologia , Compostos de Boro/efeitos adversos , Bortezomib/efeitos adversos , Ensaios Clínicos como Assunto , Gastroenteropatias/fisiopatologia , Glicina/efeitos adversos , Glicina/análogos & derivados , Humanos , Oligopeptídeos/efeitos adversos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA