Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Thromb Thrombolysis ; 49(1): 27-33, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31552545

RESUMO

A diagnosis of heparin induced thrombocytopenia (HIT) must often be made based on clinical and laboratory evidence. This was a quasi-experimental study of patients admitted from June 2016 to October 2017. The primary endpoint was the incidence of false positive results in polyspecific and IgG specific enzyme-linked immunosorbent assay (ELISA); then we compared the sensitivity and specificity of each assays in predominately cardiac patients with suspected HIT. A sensitivity/specificity analysis was conducted using serotonin release assay (SRA) as the 'gold standard'. The secondary outcome measures included length of hospital stay. We identified a total of 155 patients who met the inclusion criteria. Confirmatory tests with SRA on both groups were completed; false positive result was higher in the polyspecific group when compared to the IgG group [60% vs. 5%]. The IgG specific ELISA test yielded a sensitivity of 100% and a specificity of 95% however, the polyspecific ELISA had a low yield for specificity of 24% but maintained 100% sensitivity. In the IgG specific group with HIT-, their median length of stay was halved compared to those who were HIT + ; hospital LOS in days, IQR [30 (27-81) vs. 15 (7-33) p = 0.023] and a shorter median LOS in the ICU, IQR [24 (5-47) vs. 6 (2-14); p = 0.079]. Hospital or ICU LOS was the same in both (HIT+ and HIT-) groups managed with polyspecific ELISA. The IgG specific test had few false positive results and a high sensitivity score. Ensuring appropriate testing can bring a substantial decrease in drug expenditure, reduced length of stay and prevent unnecessary anticoagulation.


Assuntos
Cardiopatias/sangue , Cardiopatias/tratamento farmacológico , Heparina/efeitos adversos , Imunoglobulina G/sangue , Tempo de Internação , Trombocitopenia/sangue , Idoso , Ensaio de Imunoadsorção Enzimática , Feminino , Cardiopatias/epidemiologia , Heparina/administração & dosagem , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , Sensibilidade e Especificidade , Trombocitopenia/induzido quimicamente , Trombocitopenia/epidemiologia
2.
J Clin Neurosci ; 70: 37-41, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31558362

RESUMO

The inflammatory processes following traumatic brain injury (TBI) have not been fully characterized. We hypothesize that differences in systemic cytokine/chemokine (CC) levels are associated with TBI clinical outcomes. To test this hypothesis, we examined systemic levels of CCs and their relationship with patient outcomes. Plasma from acute TBI subjects was collected at 24-48 h, and the CC levels were measured using a multiplex 41-plex-kit. Clinical outcomes were assessed using the modified Rankin scale (mRS) with good outcomes defined as mRS ≤ 3 and poor outcome as mRS ≥ 4. The differences in CC concentrations between groups were then compared using the Mann-Whitney U test. Seventy-six acute TBI subjects were included in this study. In the mRS ≥ 4 group, interleukin-6 (IL-6) and interleukin-10 (IL-10) were elevated, indicating early activation of immune reaction and modulation. Simultaneously, PDGFAA and RANTES were lower in the mRS ≥ 4 group. Poor outcomes after TBI were associated with elevated levels of IL-6 and IL-10 and lower levels of PDGFAA and RANTES within 24-48 h after injury.


Assuntos
Lesões Encefálicas Traumáticas/imunologia , Inflamação/imunologia , Adulto , Lesões Encefálicas Traumáticas/complicações , Citocinas/imunologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Recuperação de Função Fisiológica/imunologia
3.
J Thromb Thrombolysis ; 47(2): 209-215, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30392138

RESUMO

Routine screening of high-risk asymptomatic trauma or surgical patients for venous thromboembolism (VTE) is controversial. Studies suggest against screening while others recognize that some patients at high risk may benefit. The purpose of this pilot study is to evaluate the benefit of routine screening using doppler ultrasonography for the early detection of deep venous thrombosis (DVT) in post-operative neurosurgical patients. This was a quasi-experimental study at a major academic tertiary care medical center. A total of 157 adults underwent cranial or spinal surgical interventions from March through August 2017 and received either standard screening (n = 104) versus routine ultrasonography screening (n = 53). There was no significant difference in incidence of DVT between the two groups: 11 (11%) in the standard screening group versus 5 (9%) in the routine screening group, p = 0.823. Upper and lower extremity ultrasonography was performed in 43 (41%) of the standard screening group versus 53 (100%) in the routine screening group, p < 0.001. DVT was identified in nearly one of every 6 ultrasonography screenings in the standard screening group versus 27 ultrasonography screenings required to identify one DVT in the routine screening group. There were the same number of screenings for upper extremity ultrasonography, but they did not yield or detect DVT; instead only superficial, untreatable, DVTs were reported. Total cost to diagnose one DVT, including screening and labor, averaged $13,664 in the standard group versus $56,525 in the routine group. Routine screening in neurosurgical patients who received VTE prophylaxis was not associated with lower incidence of VTE and mortality attributed to PE. Thus, routine screening may not be cost effective to prevent complications from DVT incidence.


Assuntos
Procedimentos Neurocirúrgicos/efeitos adversos , Embolia Pulmonar/diagnóstico , Ultrassonografia Doppler , Tromboembolia Venosa/diagnóstico por imagem , Trombose Venosa/diagnóstico por imagem , Adulto , Idoso , Análise Custo-Benefício , Feminino , Custos de Cuidados de Saúde , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , Procedimentos Neurocirúrgicos/mortalidade , Projetos Piloto , Valor Preditivo dos Testes , Embolia Pulmonar/economia , Embolia Pulmonar/mortalidade , Fatores de Risco , Texas/epidemiologia , Tromboembolia Venosa/economia , Tromboembolia Venosa/mortalidade , Trombose Venosa/economia , Trombose Venosa/mortalidade
4.
Cytokine ; 111: 334-341, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30269030

RESUMO

BACKGROUND: Unregulated inflammatory and thrombotic responses have been proposed to be important causes of early brain injury and worse clinical outcomes after subarachnoid hemorrhage (SAH). OBJECTIVE: We hypothesize that SAH is characterized by an increased inflammatory and thrombotic state and disruption of associations between these states. METHODS: This is a retrospective cohort study of 60 patients with SAH. 23 patients with unruptured aneurysms (UA) and 77 patients with traumatic brain injury (TBI) were chosen as controls. Plasma cytokine levels were measured using a 41-plex human immunoassay kit, and cytokine patterns associated with SAH, UA and TBI were identified using statistical and informatics methods. RESULTS: SAH was characterized by an increase in several cytokines and chemokines, platelet-derived factors, and growth factors. Cluster analysis identified several cytokine clusters common in SAH, UA and TBI groups - generally grouped as platelet-derived, vascular and pro-inflammatory clusters. In the UA group, the platelet-derived cluster had an inverse relationship with the inflammatory cluster which was absent in SAH. Additionally, a cluster comprising of growth and colony stimulating factors was unique to SAH. CONCLUSIONS: A cluster of cytokines involved in growth and colony stimulation was unique to SAH. Negative associations between the thrombotic and inflammatory molecules were observed in UA but not in SAH. Further studies to examine the pathophysiology behind the cluster unique to SAH and the associations between the thrombotic and inflammatory cytokines are required.


Assuntos
Citocinas/metabolismo , Inflamação/metabolismo , Hemorragia Subaracnóidea/metabolismo , Plaquetas/metabolismo , Lesões Encefálicas Traumáticas/metabolismo , Fatores Estimuladores de Colônias/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos
5.
Ann Clin Lab Sci ; 48(5): 659-664, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30373873

RESUMO

Sinonasal undifferentiated carcinoma (SNUC) is a highly malignant tumor in the nasal cavity or paranasal sinuses. Morphoproteomics has defined its biology to some degree, allowing the identification of targeted therapeutic options with clinical efficacy [1]. This study's objective was to identify putative SNUC pathways that are known to pose a block in differentiation both in early embryogenesis and in tumorigenesis or that might promote metastasis and recurrent disease. DESIGN: Morphoproteomic analysis of SNUC from a case study of this patient included immunohistochemical probes to detect c-Myc, EZH2, Sirt1 and CXCR4 protein analytes. Biomedical analytics schematically showed the interactions of these analytes with the morphoproteomic findings and illustrated targeted therapeutic options. RESULTS: Representative sections of this patient's tumor displayed plasmalemmal expression for CXCR4 and nuclear immunopositivity for c-Myc, EZH2, and Sirt1. This coincided with their block in differentiation and their proliferative state with progression into the mitotic phase. Biomedical analytics integrated the morphoproteomic findings with the undifferentiated and proliferative state of SNUC and depicted pharmacogenomic and other related factors that target the c-Myc, EZH2, Sirt1 and CXCR4 pathways. CONCLUSION: Morphoproteomics and biomedical analytics have identified c-Myc, EZH2, Sirt1 and CXCR4 pathways that collectively could contribute to the block in differentiation and increase the propensity for recurrence and metastasis in SNUC. This suggests that combinatorial therapies modulating these pathways could be used in a maintenance mode to minimize the risk of recurrent disease.


Assuntos
Carcinoma/patologia , Proteína Potenciadora do Homólogo 2 de Zeste/fisiologia , Neoplasias do Seio Maxilar/patologia , Proteômica/métodos , Proteínas Proto-Oncogênicas c-myc/fisiologia , Receptores CXCR4/fisiologia , Transdução de Sinais/fisiologia , Sirtuína 1/fisiologia , Carcinoma/química , Carcinoma/terapia , Proliferação de Células , Proteína Potenciadora do Homólogo 2 de Zeste/análise , Humanos , Neoplasias do Seio Maxilar/química , Neoplasias do Seio Maxilar/terapia , Metástase Neoplásica , Proteínas Proto-Oncogênicas c-myc/análise , Receptores CXCR4/análise , Sirtuína 1/análise
6.
Orphanet J Rare Dis ; 12(1): 181, 2017 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-29246172

RESUMO

We first introduced the concept of the mTOR pathway's involvement in congenital hyperinsulinism of infancy (CHI), based largely on morphoproteomic observations and clinical outcomes using sirolimus (rapamycin) as a therapeutic agent in infants refractory to octreotide and diazoxide treatment. Subsequent publications have verified the efficacy of such treatment in some cases but limited and variable in others. We present further evidence of a constant but variable role for the mTOR pathway in the biology of CHI and provide a strategy that allows for the short-term testing of sirolimus in individual CHI patients.


Assuntos
Hiperinsulinismo Congênito/tratamento farmacológico , Hiperinsulinismo Congênito/metabolismo , Sirolimo/uso terapêutico , Serina-Treonina Quinases TOR/metabolismo , Diazóxido/uso terapêutico , Humanos , Lactente , Octreotida/uso terapêutico , Proteômica/métodos , Transdução de Sinais/efeitos dos fármacos
7.
Ann Clin Lab Sci ; 47(4): 457-465, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28801373

RESUMO

Metformin has been proposed as a novel anti-cancer drug for adrenocortical carcinoma (ACC) based upon Poli's recent preclinical studies that 1. "in vitro" metformin modulates the ACC cell model H295R and 2. "in vivo" metformin inhibits tumor growth in a xenograft model as confirmed by a significant reduction of Ki67 [1]. Here we report on our prior clinical case study that provides proof of concept for Poli's studies. We were requested to perform morphoproteomic analysis to further define the biology of, and raise targeted therapeutic options, for a case of post-treatment and chemoresistant ACC metastatic to the liver and the lung. Profiling the patient's ACC from the liver resulted in the recommendation of metformin as a maintenance therapy, which was supported by biomedical data analysis. The patient remains on maintenance therapy with metformin and melatonin and is free of disease some 7 years post diagnosis, thus underscoring the recommendation for clinical trials employing these therapeutic agents.


Assuntos
Neoplasias do Córtex Suprarrenal/patologia , Carcinoma Adrenocortical/patologia , Biomarcadores Tumorais/metabolismo , Melatonina/uso terapêutico , Metformina/uso terapêutico , Proteômica/métodos , Neoplasias do Córtex Suprarrenal/tratamento farmacológico , Neoplasias do Córtex Suprarrenal/metabolismo , Carcinoma Adrenocortical/tratamento farmacológico , Carcinoma Adrenocortical/metabolismo , Adulto , Antioxidantes/uso terapêutico , Quimioterapia Combinada , Feminino , Humanos , Hipoglicemiantes/uso terapêutico , Imuno-Histoquímica , Prognóstico , Transdução de Sinais
8.
J Otolaryngol Head Neck Surg ; 46(1): 52, 2017 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-28818106

RESUMO

BACKGROUND: Human papillomavirus (HPV) has been identified as an etiopathogenetic factor in oropharyngeal squamous cell carcinoma. The HPV E6 and E7 oncogenes are instrumental in promoting proliferation and blocking differentiation leading to tumorigenesis. Although surgical intervention can remove such tumors, the potential for an etiologic field effect with recurrent disease is real. A downstream effector of E7 oncoprotein, enhancer of zeste homolog 2 (EZH2), is known to promote proliferation and to pose a block in differentiation and in turn, could lead to HPV-induced malignant transformation. However, the EZH2 pathway is amenable to low toxicity therapies designed to promote differentiation to a more benign state and prevent recurrent disease by inhibiting the incorporation of HPV into the genome. This is the first study using clinical specimens to demonstrate EZH2 protein expression in oropharyngeal carcinoma (OPC). METHODS: The study included eight patients with oropharyngeal carcinoma, confirmed p16INK4a- positive by immunohistochemistry (IHC). The tissue expression of E6/E7 messenger RNA (mRNA) was measured by RNAscope® in-situ hybridization technology. Expression of EZH2, Ki-67, and mitotic indices were assessed by morphoproteomic analysis. Biomedical analytics expanded the results with data from Ingenuity Pathway Analysis (IPA) and KEGG databases to construct a molecular network pathway for further insights. RESULTS: Expression of E6 and E7 oncogenes in p16INK4a- positive oropharyngeal carcinoma was confirmed. EZH2 and its correlates, including elevated proliferation index (Ki-67) and mitotic progression were also present. Biomedical analytics validated the relationship between HPV- E6 and E7 and the expression of the EZH2 pathway. CONCLUSION: There is morphoproteomic and mRNA evidence of the association of p16INK4a-HPV infection with the E6 and E7 oncogenes and the expression of EZH2, Ki-67 and mitotic progression in oropharyngeal carcinoma. The molecular network biology was confirmed by biomedical analytics as consistent with published literature. This is significant because the biology lends itself to targeted therapeutic options using metformin, curcumin, celecoxib and sulforaphane as therapeutic strategies to prevent progression or recurrence of disease.


Assuntos
Regulação Neoplásica da Expressão Gênica , Terapia de Alvo Molecular/métodos , Proteínas Oncogênicas Virais/genética , Neoplasias Orofaríngeas/virologia , Proteínas E7 de Papillomavirus/genética , Proteínas Repressoras/genética , Idoso , Biópsia por Agulha , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Feminino , Humanos , Imuno-Histoquímica , Hibridização In Situ/métodos , Masculino , Neoplasias Orofaríngeas/tratamento farmacológico , Neoplasias Orofaríngeas/patologia , Papillomaviridae/genética , Papillomaviridae/patogenicidade , Infecções por Papillomavirus/tratamento farmacológico , Infecções por Papillomavirus/genética , Infecções por Papillomavirus/patologia , Prognóstico , Proteômica , RNA Mensageiro/metabolismo , Estudos de Amostragem , Resultado do Tratamento , Infecções Tumorais por Vírus/tratamento farmacológico , Infecções Tumorais por Vírus/genética , Infecções Tumorais por Vírus/patologia
9.
Ann Clin Lab Sci ; 47(1): 3-9, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28249909

RESUMO

B-cell acute lymphoblastic leukemia (ALL) represents a malignant process in which bone marrow-derived lymphoblasts retain their undifferentiated state. Genetic testing has revealed either no identifiable cytogenetic and genomic abnormalities in such patients or a wide range of aberrations that may or may not contribute to the block in differentiation and the associated proliferation of the malignant lymphoblasts in cases of B-cell ALL. In this study, we applied morphoproteomics to a representative spectrum of cases of newly diagnosed B-cell ALL in order to identify pathways that are known to be associated with the maintenance of the undifferentiated state while promoting proliferation. Our results showed nuclear expression in a majority of the lymphoblasts from bone marrow clot preparations of each of the study cases for both silent mating type information regulation 2 homolog 1 (SIRT1), an NAD+ histone deacetylase and enhancer of Zeste homolog 2 (EZH2), a histone methyltransferase. These represent pathogenetic pathways capable of blocking differentiation and promoting proliferation of the B-cell ALL lymphoblasts. Data mining of the National Library of Medicine's MEDLINE Database and Ingenuity Pathway analysis revealed agents of relatively low toxicity-melatonin, metformin, curcumin and sulforaphane-that are capable of inhibiting directly or pharmacogenomically one or both of the SIRT1 and EZH2 pathways and should, in a combinatorial fashion, remove the block in differentiation and decrease the proliferation of the B-cell ALL lymphoblasts.


Assuntos
Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Proteômica/métodos , Transdução de Sinais , Sirtuína 1/metabolismo , Adolescente , Adulto , Criança , Pré-Escolar , Feminino , Citometria de Fluxo , Humanos , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Adulto Jovem
10.
Oncoscience ; 3(5-6): 156-63, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27489862

RESUMO

UNLABELLED: Glioblastoma multiforme (GBM) is a CNS (central nervous system) malignancy with a low cure rate. Median time to progression after standard treatment is 7 months and median overall survival is 15 months [1]. Post-treatment vasculogenesis promoted by recruitment of bone marrow derived cells (BMDCs, CD11b+ myelomonocytes) is one of main mechanisms of GBM resistance to initial chemoradiotherapy treatment [2]. Local secretion of SDF-1, cognate ligand of BMDCs CXCR4 receptors attracts BMDCs to the post-radiation tumor site.[3]. This SDF-1 hypoxia-dependent effect can be blocked by AMD3100 (plerixafor) [4]. We report a GBM case treated after chemo- radiotherapy with plerixafor and a combination of an mTOR, a Sirt1 and an EGFRvIII inhibitor. After one year temozolomide and the EGFRvIII inhibitor were stopped. Plerixafor, and the MTOR and Sirt-1 inhibitors were continued. He is in clinical and radiologic remission 30 months from the initiation of his adjuvant treatment. To our knowledge, this is the first report of a patient treated for over two years with a CXCR4 inhibitor (plerixafor), as part of his adjuvant treatment. We believe there is sufficient experimental evidence to consider AMD3100 (plerixafor) part of the adjuvant treatment of GBM. SIGNIFICANCE: The adjuvant inhibition of GBM vasculogenesis(a process different from local angiogenesis) by specifically blocking the migration of BMDCs to the primary tumor site with inhibitors of the CXCR4/SDF-1 axis represents a potential novel therapeutic approach to GBM. There is significant pre-clinical evidence and validation for its use as demonstrated in a patient derived tumor xenograft model of GBM. Together with other specific anti-tumoral therapies, the active inhibition of vasculogenesis in the adjuvant treatment of GBM is deserving of further exploration.

11.
Oncotarget ; 7(27): 41612-41621, 2016 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-27223432

RESUMO

BACKGROUND: It has been proposed that resistance to rapalog therapies in renal cell carcinoma (RCC) is due to adaptive switching from mammalian target of rapamycin complex 1 (mTORC1) to mTORC2. OBJECTIVE: To combine phosphoprotein staining and applied biomedical analytics to investigate resistance signatures in patients with metastatic RCC progressing on rapalog therapies. DESIGN: We applied morphoproteomic analysis to biopsy specimens from nine patients with metastatic RCC who continued to show clinical progression of their tumors while being treated with a rapalog. RESULTS: In patients who were on temsirolimus or everolimus at the time of biopsy, a moderate to strong expression of phosphorylated (p)-mTOR (Ser 2448) in the nuclear compartment with concomitant expression of p-Akt (Ser 473) confirmed the mTORC2 pathway. Concomitant moderate to strong nuclear expression of p-ERK 1/2 (Thr202/Tyr204) and p-STAT3 (Tyr705) was confirmed. Histopathologic changes of hypoxic-type coagulative necrosis in 5 cases as well as identification of insulin-like growth factor-1 receptor (IGF-1R) expression and histone methyltransferase EZH2 in all tumors studied suggested that hypoxia also contributed to the resistance signature. Biomedical analytics provided insight into therapeutic options that could target such adaptive and pathogenetic mechanisms. CONCLUSIONS: Morphoproteomics and biomedical analytics confirm mTORC2/Akt as a resistance signature to rapalog therapy in metastatic RCC and demonstrate activation of the prosurvival ERK and STAT3 pathways and involvement of hypoxic pathways that contribute to pathogenesis of such adaptive resistance. These results highlight the need for a novel combinatorial therapeutic approach in metastatic RCC progressing on rapalogs.


Assuntos
Carcinoma de Células Renais , Resistencia a Medicamentos Antineoplásicos , Neoplasias Renais , Proteômica/métodos , Transdução de Sinais/efeitos dos fármacos , Sirolimo/análogos & derivados , Apoptose/efeitos dos fármacos , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Tamanho Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Biologia Computacional/métodos , Progressão da Doença , Ativação Enzimática/efeitos dos fármacos , Everolimo/uso terapêutico , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Perfilação da Expressão Gênica/métodos , Humanos , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , Metástase Neoplásica , Proteína Oncogênica v-akt/metabolismo , Fator de Transcrição STAT3/metabolismo , Sirolimo/uso terapêutico , Carga Tumoral/efeitos dos fármacos
12.
Ann Clin Lab Sci ; 45(6): 692-701, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26663801

RESUMO

NUT midline carcinoma is a rare entity arising primarily in the midline of teenagers and young adults. Genomically, it is associated with a translocation involving a nuclear protein in testis (NUT) gene with other genes, most commonly, the BRD4 gene. The resultant is a partial or near total block in differentiation of tumor cells into mature squamous elements. Such tumors are resistant to conventional therapy with a reported mean survival at less than 1 year. In this study, we investigated two cases with genomic confirmation as NUT midline carcinoma by morphoproteomic analysis using immunohistochemical antibodies. Our results showed overexpression, largely in the undifferentiated cells of the tumors of: 1) Stemness marker, SRY (sex determining region Y)-box 2 (Sox2); 2) Constitutive activation of the mTORC2 pathway with expression of total insulin-like growth factor-1 receptor (IGF-1R[Tyr1165/1166]), and nuclear p-mTOR (Ser 2448) and p-Akt (Ser 473); and 3) c-Myc, silent mating type information regulation 2 homolog 1 (Sirt1) and histone methyltransferase enhancer of Zeste, Drosophila, homolog 2 (EZH2) as molecular impediments to differentiation. These data were analyzed through the use of QIAGEN's Ingenuity(®) Pathway Analysis (IPA(®), QIAGEN Redwood City, www.qiagen.com/ingenuity). The results established the interconnection of these pathways and molecules, and identified several pharmacogenomic agents--melatonin, metformin, vorinostat, curcumin, and sulforaphane--that have the potential to remove the block in differentiation and lead to the establishment of a more benign form of NUT midline carcinoma.


Assuntos
Carcinoma/genética , Carcinoma/patologia , Neoplasias do Mediastino/genética , Neoplasias do Mediastino/patologia , Proteínas Nucleares/genética , Proteínas Oncogênicas/genética , Adolescente , Adulto , Carcinoma/metabolismo , Carcinoma/terapia , Proteína Potenciadora do Homólogo 2 de Zeste , Feminino , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Isotiocianatos/farmacologia , Masculino , Neoplasias do Mediastino/metabolismo , Neoplasias do Mediastino/terapia , Metformina/farmacologia , Terapia de Alvo Molecular/métodos , Proteínas de Neoplasias , Proteínas Nucleares/metabolismo , Proteínas Oncogênicas/metabolismo , Complexo Repressor Polycomb 2/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Sirtuína 1/metabolismo , Sulfóxidos , Serina-Treonina Quinases TOR/metabolismo
13.
Oncotarget ; 6(38): 40642-54, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26510912

RESUMO

BACKGROUND: Despite advances in molecular medicine over recent decades, there has been little advancement in the treatment of osteosarcoma. We performed comprehensive molecular profiling in two cases of metastatic and chemotherapy-refractory osteosarcoma to guide molecularly targeted therapy. PATIENTS AND METHODS: Hybridization capture of >300 cancer-related genes plus introns from 28 genes often rearranged or altered in cancer was applied to >50 ng of DNA extracted from tumor samples from two patients with recurrent, metastatic osteosarcoma. The DNA from each sample was sequenced to high, uniform coverage. Immunohistochemical probes and morphoproteomics analysis were performed, in addition to fluorescence in situ hybridization. All analyses were performed in CLIA-certified laboratories. Molecularly targeted therapy based on the resulting profiles was offered to the patients. Biomedical analytics were performed using QIAGEN's Ingenuity® Pathway Analysis. RESULTS: In Patient #1, comprehensive next-generation exome sequencing showed MET amplification, PIK3CA mutation, CCNE1 amplification, and PTPRD mutation. Immunohistochemistry-based morphoproteomic analysis revealed c-Met expression [(p)-c-Met (Tyr1234/1235)] and activation of mTOR/AKT pathway [IGF-1R (Tyr1165/1166), p-mTOR [Ser2448], p-Akt (Ser473)] and expression of SPARC and COX2. Targeted therapy was administered to match the P1K3CA, c-MET, and SPARC and COX2 aberrations with sirolimus+ crizotinib and abraxane+ celecoxib. In Patient #2, aberrations included NF2 loss in exons 2-16, PDGFRα amplification, and TP53 mutation. This patient was enrolled on a clinical trial combining targeted agents temsirolimus, sorafenib and bevacizumab, to match NF2, PDGFRα and TP53 aberrations. Both the patients did not benefit from matched therapy. CONCLUSIONS: Relapsed osteosarcoma is characterized by complex signaling and drug resistance pathways. Comprehensive molecular profiling holds great promise for tailoring personalized therapies for cancer. Methods for such profiling are evolving and need to be refined to better assist clinicians in making treatment decisions based on the large amount of data that results from this type of testing. Further research in this area is warranted.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Ósseas/genética , Perfilação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Terapia de Alvo Molecular , Osteossarcoma/genética , Medicina de Precisão , Adolescente , Adulto , Antineoplásicos/uso terapêutico , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/patologia , Humanos , Técnicas Imunoenzimáticas , Masculino , Osteossarcoma/tratamento farmacológico , Osteossarcoma/patologia , Proteômica , Adulto Jovem
14.
Oncotarget ; 5(1): 95-102, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24395633

RESUMO

UNLABELLED: Although Hodgkin's lymphoma (HL) was one of the first human cancers to be cured by chemotherapy, no new agents other than brentuximab vedotin (Adcetris®, CD 30 directed antibody drug conjugate) have received US Food and Drug Administration (FDA) approval for HL since 1977. Subsets of young adult patients with HL continue to relapse, even after stem cell transplantation, warranting new approaches. Against this background, we report a dramatic response in a young patient with advanced HL refractory to the standard treatment who responded to the combination of a pan-histone deacetylase inhibitor (vorinostat, suberoylanilide hydroxamic acid, SAHA) and mammalian target of rapamycin (mTOR) inhibitor therapy (sirolimus,rapamume). In-depth immunohistochemical and morphoproteomic analyses of this exceptional responder to targeted therapy have yielded potential insights into the biology of advanced HL. The PI3K/AKT/mTOR pathway is a commonly activated pathway in multiple tumor types including HL. The patient was treated using therapy based on mechanistic in vitro data demonstrating that combined histone deacetylase (HDAC) and mTOR inhibition act together on this pathway, resulting in inhibition of reciprocal feedback networks, leading to better anti-proliferative activity. The in vivo response signature from this patient's tissue sample sheds light on immune dysregulation in HL. We describe the response signature achieved from targeting immune dysregulation in addition to targeting the key oncogenic PI3K/AKT/mTOR pathway. We also expand on the role of rapamycin analogs in oncology. This study supports a role for an immune-type pathogenesis that is amenable to immune modulating targeted therapy in refractory HL. SIGNIFICANCE: We report an exceptional responder to molecularly targeted and immune modulator therapy in advanced Hodgkin's lymphoma. The morphoproteomic/morphometric findings in this "unusual responder" patient's relapsed HL that correlate best, as a response signature with the subsequent clinical remission following rapamycin (sirolimus) and vorinostat (SAHA) therapies, center on an immune dysregulation involving an imbalance between effector and functional T regulatory cells in addition to targeting the mTOR pathway. This underscores the need for an approach illustrated in our study--namely of focusing on pathogenetic mechanisms and combinatorial therapies that target both the pathogenesis and adaptive responses to contemplated therapies.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Doença de Hodgkin/tratamento farmacológico , Adulto , Feminino , Doença de Hodgkin/imunologia , Doença de Hodgkin/patologia , Humanos , Ácidos Hidroxâmicos/administração & dosagem , Fatores Imunológicos/administração & dosagem , Terapia de Alvo Molecular , Inibidores de Fosfoinositídeo-3 Quinase , Sirolimo/administração & dosagem , Sirolimo/análogos & derivados , Serina-Treonina Quinases TOR/antagonistas & inibidores , Vorinostat
15.
Cancer Res ; 73(20): 6111-7, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-23955390

RESUMO

Although many clinicians and researchers work to understand cancer, there has been limited success to effectively combine forces and collaborate over time, distance, data, and budget constraints. Here we present a workflow template for multidisciplinary cancer therapy that was developed during the 2nd Annual Workshop on Cancer Systems Biology sponsored by Tufts University, Boston, Massachusetts, in July 2012. The template was applied to the development of a metronomic therapy backbone for neuroblastoma. Three primary groups were identified: clinicians, biologists, and quantitative scientists (mathematicians, computer scientists, and engineers). The workflow described their integrative interactions; parallel or sequential processes; data sources and computational tools at different stages as well as the iterative nature of therapeutic development from clinical observations to in vitro, in vivo, and clinical trials. We found that theoreticians in dialog with experimentalists could develop calibrated and parameterized predictive models that inform and formalize sets of testable hypotheses, thus speeding up discovery and validation while reducing laboratory resources and costs. The developed template outlines an interdisciplinary collaboration workflow designed to systematically investigate the mechanistic underpinnings of a new therapy and validate that therapy to advance development and clinical acceptance.


Assuntos
Oncologia/organização & administração , Neoplasias/terapia , Fluxo de Trabalho , Ensaios Clínicos como Assunto/métodos , Gerenciamento Clínico , Humanos , Oncologia/métodos
16.
Ann Clin Lab Sci ; 42(3): 243-57, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22964612

RESUMO

BACKGROUND: Hypoxia (3 to 5% oxygen) is essential in maintaining the plasticity of embryonic stem cells and permitting their transformation via epithelial-mesenchymal transition(EMT) and mesenchymal-epithelial transition(MET) into tissues and organs of the developing fetus. Similarly, a relatively hypoxic microenvironment supports the development of tumor cells with stemness and epithelial-mesenchymal properties and capabilities. At the same time, such adaptation results in the tumor cells becoming relatively resistant to chemotherapy and radiation therapy and promotes intravasation into blood vessels with metastasis. In this context, current therapeutic strategies designed to target tumoral angiogenesis could promote stemness and EMT by rendering tumor cells more hypoxic, leading to chemoradioresistance and metastatic and recurrent disease. OBJECTIVE: The purpose of this report is to present a conceptual model that illustrates the impact of an hypoxic microenvironment on the signal transduction pathways involved in the hypoxia pathway. We will show the molecular connectivity and correlative association of these pathways with protein analytes in both embryogenesis and oncogenesis in order to strengthen our hypothesis that oncogenesis recapitulates embryogenesis. Finally, we propose to use the model as a basis for the construction of combinatorial, therapeutic options from existing pharmaceutical and nutraceutical agents that may obviate tumoral adaptation to hypoxia. METHODS: Morphoproteomics and biomedical analytics. APPLICATION AND RESULTS: Archival data retrieved from morphoproteomic analysis of glioblastoma multiforme(GBM) cases revealed proteomic correlates of tumoral necrosis and associated hypoxia pathway signaling. Biomedical analytics using Ingenuity Pathway Analysis (IPA) showed comparative validation of the hypoxia pathway, as demonstrated by morphoproteomics in GBM, both with the hypoxia-induced genes in neuroblastoma and with the networks associated with embryogenesis. Additionally, therapeutic agents known to have activity against various components of the hypoxia pathway (identified by morphoproteomic analysis in GBM) were validated using UNIPROT identifiers entered into IPA and Path Designer. These therapies also connected with the hypoxia signature in neuroblastoma and embryogenesis. CONCLUSION: The application of morphoproteomics to define the presence of an adaptive hypoxia pathway in GBM accords with biomedical analytics in the demonstration of concordant interaction with the hypoxia signature in neuroblastoma and embryogenesis, providing proof of concept that oncogenesis recapitulates embryogenesis. This approach also validates a new combinatorial therapeutic strategy targeting the hypoxia pathway and designed to prevent tumoral adaptation, chemoradioresistance and recurrent disease.


Assuntos
Tecnologia Biomédica/métodos , Transformação Celular Neoplásica/patologia , Desenvolvimento Embrionário , Proteômica/métodos , Transdução de Sinais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Hipóxia Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Mineração de Dados , Bases de Dados como Assunto , Desenvolvimento Embrionário/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Coloração e Rotulagem , Serina-Treonina Quinases TOR/metabolismo
17.
J Trauma Acute Care Surg ; 72(1): 112-8, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22310124

RESUMO

BACKGROUND: Trauma centers are caring for increased proportions of elderly patients. Although age and Injury Severity Score are independently associated with mortality, trauma centers were originally designed to care for seriously injured patients without age-specific guidelines. We hypothesized that elderly patients would have different complication patterns than their younger counterparts. METHODS: The trauma registry of an American College of Surgeons -verified Level I trauma center was queried for all patients older than 14 years admitted between January 2005 and December 2008. Mechanism, mortality, and complications were evaluated after dividing patients into eight age groups. RESULTS: Of the 15,223 patients, 13% were elderly (≥65), and 86% were injured via a blunt mechanism. Increasing age correlated with fatality (all Injury Severity Scores), end-organ failure, and thromboembolic complications (deep venous thrombosis and coagulopathy). Analysis revealed a significant breakpoint at 45 years of age for mortality, decubitus ulcer, and renal failure (all p values <0.05). Infectious complications (sepsis, wound infection, and abscess) all peaked between 45 years and 65 years and then declined with increasing age. CONCLUSIONS: We document that elderly trauma patients suffer the same complications as their younger counterparts, albeit at a different rate. More importantly, we identified a "breakpoint" of increased risk of complications and mortality at greater than 45 years. Although the mechanisms behind these observations remain unknown, understanding their unique patterns may allow appropriate allocation of resources and focus research efforts on interventions that should improve outcomes.


Assuntos
Centros de Traumatologia/estatística & dados numéricos , Ferimentos e Lesões/complicações , Adolescente , Adulto , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Escala de Gravidade do Ferimento , Tempo de Internação , Masculino , Pessoa de Meia-Idade , Fatores de Risco , Texas/epidemiologia , Ferimentos e Lesões/mortalidade , Adulto Jovem
18.
J Biomed Inform ; 45(2): 372-87, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22200681

RESUMO

MOTIVATION: Although trauma is the leading cause of death for those below 45years of age, there is a dearth of information about the temporal behavior of the underlying biological mechanisms in those who survive the initial trauma only to later suffer from syndromes such as multiple organ failure. Levels of serum cytokines potentially affect the clinical outcomes of trauma; understanding how cytokine levels modulate intra-cellular signaling pathways can yield insights into molecular mechanisms of disease progression and help to identify targeted therapies. However, developing such analyses is challenging since it necessitates the integration and interpretation of large amounts of heterogeneous, quantitative and qualitative data. Here we present the Pathway Semantics Algorithm (PSA), an algebraic process of node and edge analyses of evoked biological pathways over time for in silico discovery of biomedical hypotheses, using data from a prospective controlled clinical study of the role of cytokines in multiple organ failure (MOF) at a major US trauma center. A matrix algebra approach was used in both the PSA node and PSA edge analyses with different matrix configurations and computations based on the biomedical questions to be examined. In the edge analysis, a percentage measure of crosstalk called XTALK was also developed to assess cross-pathway interference. RESULTS: In the node/molecular analysis of the first 24h from trauma, PSA uncovered seven molecules evoked computationally that differentiated outcomes of MOF or non-MOF (NMOF), of which three molecules had not been previously associated with any shock/trauma syndrome. In the edge/molecular interaction analysis, PSA examined four categories of functional molecular interaction relationships--activation, expression, inhibition, and transcription--and found that the interaction patterns and crosstalk changed over time and outcome. The PSA edge analysis suggests that a diagnosis, prognosis or therapy based on molecular interaction mechanisms may be most effective within a certain time period and for a specific functional relationship.


Assuntos
Algoritmos , Progressão da Doença , Insuficiência de Múltiplos Órgãos/metabolismo , Insuficiência de Múltiplos Órgãos/patologia , Choque Traumático/patologia , Transdução de Sinais , Humanos , Semântica
19.
J Investig Med ; 59(6): 893-903, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21712727

RESUMO

Today, there is an ever-increasing amount of biological and clinical data available that could be used to enhance a systems-based understanding of disease progression through innovative computational analysis. In this article, we review a selection of published research regarding computational methods, primarily from systems biology, which support translational research from the molecular level to the bedside, with a focus on applications in trauma and critical care. Trauma is the leading cause of mortality in Americans younger than 45 years, and its rapid progression offers both opportunities and challenges for computational analysis of trends in molecular patterns associated with outcomes and therapeutic interventions.This review presents methods and domain-specific examples that may inspire the development of new algorithms and computational methods that use both molecular and clinical data for diagnosis, prognosis, and therapy in disease progression.


Assuntos
Biologia Computacional/métodos , Pesquisa Translacional Biomédica/métodos , Adulto , Algoritmos , Teorema de Bayes , Pesquisa Biomédica , Simulação por Computador , Progressão da Doença , Humanos , Pessoa de Meia-Idade , Modelos Estatísticos , Prognóstico , Biologia de Sistemas , Teoria de Sistemas , Resultado do Tratamento , Estados Unidos , Ferimentos e Lesões
20.
J Am Coll Surg ; 209(3): 320-31, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19717036

RESUMO

BACKGROUND: Shock is a prime inciting event for postinjury multiple organ failure (MOF), believed to induce a state of injurious systemic inflammation. In animal models of hemorrhagic shock, early (< 24 hours) changes in cytokine production are an index of the systemic inflammatory response syndrome. However, their predictive value in trauma patients remains to be fully elucidated. STUDY DESIGN: In a prospective observational pilot study of > 1 year at an urban Level I trauma center, serial (every 4 hours) serum cytokine levels were determined during a 24-hour period using multiplex suspension immunoassay in patients with major torso trauma (excluding severe brain injury) who met criteria for standardized shock resuscitation. Temporal cytokine expression was assessed during shock resuscitation in severe trauma patients to predict risk for MOF. MOF was assessed with the Denver score. RESULTS: Of 48 study patients (mean age 39 +/- 3 years, 67% men, 88% blunt mechanism, mean Injury Severity Score 25 +/- 2), MOF developed in 11 (23%). MOF patients had a considerably higher mortality (64% versus 3%) and fewer ICU-free days (3.5 +/- 2 versus 17.8 +/- 1.3 days) compared with non-MOF patients. Traditional predictors of MOF, including age (45 +/- 7 versus 38 +/- 3 years; p=0.21), Injury Severity Score (26 +/- 3 versus 25 +/- 2; p=0.67), admission hemoglobin (11.4 +/- 0.9 versus 12.1 +/- 0.5 g/dL; p=0.22), international normalized ratio (1.6 +/- 0.2 versus 1.4 +/- 0.06; p=0.17), and base deficit (9.0 +/- 2 versus 7.1 +/- 0.8; p=0.19), were not significantly different between MOF and non-MOF patients. Statistical analysis identified six candidate predictors of MOF: inducible protein 10, macrophage inflammatory protein-1beta, interleukin-10, interleukin-6, interleukin-1Ra, and eotaxin. CONCLUSIONS: These data provide insight into cytokine expression during traumatic shock that can enable earlier identification of patients at risk for development of MOF.


Assuntos
Citocinas/sangue , Insuficiência de Múltiplos Órgãos/classificação , Choque/sangue , Adulto , Teorema de Bayes , Feminino , Humanos , Imunoensaio/métodos , Escala de Gravidade do Ferimento , Masculino , Pessoa de Meia-Idade , Traumatismo Múltiplo , Projetos Piloto , Valor Preditivo dos Testes , Estudos Prospectivos , Medição de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA