1
|
Berlow NE, Grasso CS, Quist MJ, Cheng M, Gandour-Edwards R, Hernandez BS, Michalek JE, Ryan C, Spellman P, Pal R, Million LS, Renneker M, Keller C. Deep Functional and Molecular Characterization of a High-Risk Undifferentiated Pleomorphic Sarcoma. Sarcoma 2020; 2020:6312480. [PMID: 32565715 PMCID: PMC7285280 DOI: 10.1155/2020/6312480] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/07/2020] [Accepted: 02/10/2020] [Indexed: 11/29/2022] Open
Abstract
Nonrhabdomyosarcoma soft-tissue sarcomas (STSs) are a class of 50+ cancers arising in muscle and soft tissues of children, adolescents, and adults. Rarity of each subtype often precludes subtype-specific preclinical research, leaving many STS patients with limited treatment options should frontline therapy be insufficient. When clinical options are exhausted, personalized therapy assignment approaches may help direct patient care. Here, we report the results of an adult female STS patient with relapsed undifferentiated pleomorphic sarcoma (UPS) who self-drove exploration of a wide array of personalized Clinical Laboratory Improvement Amendments (CLIAs) level and research-level diagnostics, including state of the art genomic, proteomic, ex vivo live cell chemosensitivity testing, a patient-derived xenograft model, and immunoscoring. Her therapeutic choices were also diverse, including neoadjuvant chemotherapy, radiation therapy, and surgeries. Adjuvant and recurrence strategies included off-label and natural medicines, several immunotherapies, and N-of-1 approaches. Identified treatment options, especially those validated during the in vivo study, were not introduced into the course of clinical treatment but did provide plausible treatment regimens based on FDA-approved clinical agents.
Collapse
Affiliation(s)
- Noah E. Berlow
- Children's Cancer Therapy Development Institute, Beaverton, OR 97005, USA
- Electrical and Computer Engineering, Texas Tech University, Lubbock, TX 79409, USA
- Division of Hematology-Oncology, University of California, Los Angeles, CA 90095, USA
| | - Catherine S. Grasso
- Division of Hematology-Oncology, University of California, Los Angeles, CA 90095, USA
| | - Michael J. Quist
- Division of Hematology-Oncology, University of California, Los Angeles, CA 90095, USA
| | | | - Regina Gandour-Edwards
- Department of Pathology & Laboratory Medicine, UC Davis Health System, Sacramento, CA 95817, USA
| | - Brian S. Hernandez
- Department of Epidemiology and Biostatistics, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA
| | - Joel E. Michalek
- Department of Epidemiology and Biostatistics, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA
| | - Christopher Ryan
- School of Medicine, Oregon Health and Science University, Portland, OR 97239, USA
| | - Paul Spellman
- School of Medicine, Oregon Health and Science University, Portland, OR 97239, USA
| | - Ranadip Pal
- Electrical and Computer Engineering, Texas Tech University, Lubbock, TX 79409, USA
| | - Lynn S. Million
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Mark Renneker
- Patient-Directed Consultations, San Francisco, CA 94116, USA
- Department of Family Medicine, University of California San Francisco, San Francisco, CA 94143, USA
| | - Charles Keller
- Children's Cancer Therapy Development Institute, Beaverton, OR 97005, USA
- Electrical and Computer Engineering, Texas Tech University, Lubbock, TX 79409, USA
| |
Collapse
|
2
|
Groisberg R, Roszik J, Conley AP, Lazar AJ, Portal DE, Hong DS, Naing A, Herzog CE, Somaiah N, Zarzour MA, Patel S, Brown RE, Subbiah V. Genomics, Morphoproteomics, and Treatment Patterns of Patients with Alveolar Soft Part Sarcoma and Response to Multiple Experimental Therapies. Mol Cancer Ther 2020; 19:1165-1172. [PMID: 32127467 DOI: 10.1158/1535-7163.mct-19-0579] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 12/18/2019] [Accepted: 02/17/2020] [Indexed: 01/18/2023]
Abstract
Overexpression of transcription factor 3 in alveolar soft part sarcoma(ASPS) results in upregulation of cell proliferation pathways. No standard treatment algorithm exists for ASPS; multikinase inhibitors[tyrosine kinase inhibitor (TKI)] and immune checkpoint inhibitors (ICI) have shown clinical benefit. To date, no studies have reported on management strategies or sequencing of therapy. We evaluated ASPS treatment patterns and responses in an experimental therapeutics clinic. Genomic and morphoproteomic analysis was performed to further elucidate novel targets. We retrospectively reviewed patients with ASPS treated on clinical trials. Demographic and clinical next-generation sequencing (NGS) profiles were collected. AACR GENIE database was queried to further evaluate aberrations in ASPS. Morphoproteomic analysis was carried out to better define the biology of ASPS with integration of genomic and proteomic findings. Eleven patients with ASPS were identified; 7 received NGS testing and mutations in CDKN2A (n = 1) and hepatocyte growth factor (n = 1) were present. Ten patients were treated with TKIs with stable disease as best response and 4 patients with ICI (three partial responses). Within GENIE, 20 patients were identified harboring 3 called pathogenic mutations. Tumor mutation burden was low in all samples. Morphoproteomic analysis confirmed the expression of phosphorylated c-Met. In addition, fatty acid synthase and phosphorylated-STAT3 were detected in tumor cell cytoplasm and nuclei. Patients with ASPS have a quiescent genome and derive clinical benefit from VEGF-targeting TKIs. Morphoproteomic analysis has provided both additional correlative pathways and angiogenic mechanisms that are targetable for patients with ASPS. Our study suggests that sequential therapy with TKIs and immune checkpoint inhibitors is a reasonable management strategy.
Collapse
Affiliation(s)
- Roman Groisberg
- Department of Melanoma/Sarcoma Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Jason Roszik
- Department of Melanoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anthony P Conley
- Department of Sarcoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alexander J Lazar
- Department of Anatomic Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Daniella E Portal
- Department of Melanoma/Sarcoma Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - David S Hong
- Department of Investigational Cancer Therapeutics (Phase 1 Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Aung Naing
- Department of Investigational Cancer Therapeutics (Phase 1 Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cynthia E Herzog
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Neeta Somaiah
- Department of Sarcoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Maria A Zarzour
- Department of Sarcoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shreyaskumar Patel
- Department of Sarcoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Robert E Brown
- Department of Pathology and Laboratory Medicine, UTHealth McGovern Medical School, Houston, Texas
| | - Vivek Subbiah
- Department of Investigational Cancer Therapeutics (Phase 1 Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas. .,Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
3
|
Wang L, Zhan Y, Wu Z, Lin M, Jin X, Jiang L, Qiu Y. A novel multitarget kinase inhibitor BZG with potent anticancer activity in vitro and vivo enhances efficacy of sorafenib through PI3K pathways in hepatocellular carcinoma cells. Biomed Pharmacother 2020; 125:110033. [PMID: 32187962 DOI: 10.1016/j.biopha.2020.110033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 02/14/2020] [Accepted: 02/17/2020] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES BZG as a novel multitarget kinase inhibitor, has been proved to inhibit the proliferation of hepatocellular carcinoma (HCC) previously. In this study, we aimed at investigating the underlying mechanisms of BZG with and without sorafenib and evaluating their anti-tumor effects as well as whether BZG could inhibit the activation of phosphoinositide 3-kinase (PI3K)/AKT signaling which is associated with acquired resistance to sorafenib. METHODS We evaluated the proliferation of HCC cells by CCK-8 assay and colony formation assay. Cell apoptosis was assessed by Hoechst 33342 staining assay and flow cytometry. Western blot was used to detect the critical enzymes in the PI3K pathways and the expression of p-ERK after BZG alone and combined with sorafenib treatments. Huh-7 hepatocellular carcinoma xenograft model was used to evaluate the anti-carcinoma effects of BZG alone and in combination in vivo. HE staining and TUNEL assay tested the necrosis of tumor tissue and apoptosis of tumor cells. RESULTS BZG could inhibit the proliferation of HCC cells in a dose-dependent manner. The combination of BZG and sorafenib produced synergistic effects. PI3K and p-ERK pathway were involved in the anti-tumor functions of BZG alone and when combined with sorafenib. In addition, the combination treatment was seen to be more effective in inhibiting the expression of p-AKT, p-ERK and p-mTOR. Furthermore, Tumor necrosis and cell apoptosis were also observed in Huh-7 hepatocellular carcinoma xenograft models. CONCLUSIONS BZG is an attractive agent for treating HCC. The effects of BZG and sorafenib's co-treatment on HCC are more effective than BZG or sorafenib alone.
Collapse
Affiliation(s)
- Li Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang, 310000, PR China
| | - Yaqiong Zhan
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang, 310000, PR China
| | - Zhe Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang, 310000, PR China
| | - Mengjia Lin
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang, 310000, PR China
| | - Xuehang Jin
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang, 310000, PR China
| | - Lushun Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang, 310000, PR China
| | - Yunqing Qiu
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang, 310000, PR China.
| |
Collapse
|
4
|
Preclinical efficacy of a novel dual PI3K/mTOR inhibitor, CMG002, alone and in combination with sorafenib in hepatocellular carcinoma. Cancer Chemother Pharmacol 2019; 84:809-817. [PMID: 31385002 DOI: 10.1007/s00280-019-03918-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 07/29/2019] [Indexed: 12/23/2022]
Abstract
PURPOSE Sorafenib has been the only first systemic drug that improves survival of patients with advanced hepatocellular carcinoma (HCC). However, because the response rate of sorafenib is relatively low, novel therapeutic strategies are needed to improve survival in patients with HCC. This study investigated the effect of CMG002 alone and in combination with sorafenib on HCC in vitro and vivo. METHODS The effect of a newly developed dual PI3K/mTOR inhibitor, CMG002, on the proliferation of Huh-7 and HepG2 HCC cells was investigated using the MTT assay. Western blotting was performed to assess phosphorylation of the key enzymes in the Ras/Raf/MAPK and PI3K/AKT/mTOR pathways. HepG2 cells were inoculated into mice, which were treated with vehicle, sorafenib, CMG002, and their combinations. Tumor cell proliferation and tumor angiogenesis were evaluated by immunohistochemical analysis of Ki-67 and CD31, respectively. Tumor cell apoptosis was detected by the terminal deoxynucleotidyl transferase dUTP nick end labeling assay. Levels of key enzymes in the Ras/Raf/MAPK and PI3K/AKT/mTOR pathways were evaluated by western blot analysis. RESULTS The combination of sorafenib and CMG002 additively inhibited Huh-7 and HepG2 cell proliferation compared to single-agent treatment. Sorafenib and CMG002 as single agents differentially inhibited or activated key enzymes in the Ras/Raf/MAPK and PI3K/AKT/mTOR pathways. The combination of sorafenib and CMG002 inhibited all key enzymes in the two pathways. Treatment with CMG002 for 4 weeks alone and in combination with sorafenib strongly inhibited tumor growth. CMG002 inhibited HCC cell proliferation, induced apoptosis, and decreased tumor angiogenesis. Furthermore, these effects were enhanced when CMG002 was combined with sorafenib. CONCLUSIONS The combination of CMG002 and sorafenib significantly inhibited HCC cell proliferation and tumorigenesis by inhibiting the Ras/Raf/MAPK and PI3K/AKT/mTOR pathways. These findings suggest that CMG002 to be a potential novel candidate treatment for HCC.
Collapse
|
5
|
Brown RE, Buryanek J, Katz AM, Paz K, Wolff JE. Alveolar rhabdomyosarcoma: morphoproteomics and personalized tumor graft testing further define the biology of PAX3-FKHR(FOXO1) subtype and provide targeted therapeutic options. Oncotarget 2018; 7:46263-46272. [PMID: 27323832 PMCID: PMC5216796 DOI: 10.18632/oncotarget.10089] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 06/03/2016] [Indexed: 12/13/2022] Open
Abstract
Alveolar rhabdomyosarcoma (ARMS) represents a block in differentiation of malignant myoblasts. Genomic events implicated in the pathogenesis of ARMS involve PAX3-FKHR (FOXO1) or PAX7-FKHR (FOXO1) translocation with corresponding fusion transcripts and fusion proteins. Commonalities in ARMS include uncontrollable proliferation and failure to differentiate. The genomic-molecular correlates contributing to the etiopathogenesis of ARMS incorporate PAX3-FKHR (FOXO1) fusion protein stimulation of the IGF-1R, c-Met and GSK3-β pathways. With sequential morphoproteomic profiling on such a case in conjunction with personalized tumor graft testing, we provide an expanded definition of the biology of PAX3-FKHR (FOXO1) ARMS that integrates genomics, proteomics and pharmacogenomics. Moreover, therapies that target the genomic and molecular biology and lead to tumoral regression and/or tumoral growth inhibition in a xenograft model of ARMS are identified.
Collapse
Affiliation(s)
- Robert E Brown
- Department of Pathology & Laboratory Medicine, UT Health, McGovern Medical School, Houston, TX 77025, USA
| | - Jamie Buryanek
- Department of Pathology & Laboratory Medicine, UT Health, McGovern Medical School, Houston, TX 77025, USA
| | - Amanda M Katz
- Scientific Operations, Champions Oncology, Baltimore, MD 21205, USA
| | - Keren Paz
- Scientific Operations, Champions Oncology, Baltimore, MD 21205, USA
| | - Johannes E Wolff
- Present address: Novartis Pharmaceuticals Corporation, East Hanover, NJ 07936, USA
| |
Collapse
|
6
|
Brown RE, Buryanek J, Tammisetti VS, McGuire MF, Csencsits-Smith K. Morphoproteomics and biomedical analytics confirm the mTORC2/Akt pathway as a resistance signature and activated ERK and STAT3 as concomitant prosurvival/antiapoptotic pathways in metastatic renal cell carcinoma (RCC) progressing on rapalogs: pathogenesis and therapeutic options. Oncotarget 2018; 7:41612-41621. [PMID: 27223432 PMCID: PMC5173082 DOI: 10.18632/oncotarget.9508] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 04/16/2016] [Indexed: 11/25/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Robert E Brown
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston McGovern Medical School, TX 77030, Houston, USA
| | - Jamie Buryanek
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston McGovern Medical School, TX 77030, Houston, USA
| | - Varaha S Tammisetti
- Diagnostic and Interventional Imaging, The University of Texas Health Science Center at Houston McGovern Medical School, TX 77030, Houston, USA
| | - Mary F McGuire
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston McGovern Medical School, TX 77030, Houston, USA
| | - Keri Csencsits-Smith
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston McGovern Medical School, TX 77030, Houston, USA
| |
Collapse
|
7
|
Abstract
The management of choroid plexus carcinoma (CPC) is challenging and multifaceted. Here, we discuss a 3-year-old girl with CPC and Li-Fraumeni syndrome who achieved full remission after surgery and chemotherapy, with radiation therapy spared. At recurrence, we used a novel, standard-dose cytotoxic chemotherapy regimen, focal proton radiation therapy, and targeted agents based on morphoproteomic analysis to achieve long-term survival. We highlight the rationale for our therapy at recurrence, as well as the risk-benefit analyses necessary in decision making for these patients. Our strategy may be effective in managing other patients with recurrent CPC and Li-Fraumeni syndrome.
Collapse
|
8
|
Brown RE, Naqvi S, McGuire MF, Buryanek J, Karni RJ. Morphoproteomics, E6/E7 in-situ hybridization, and biomedical analytics define the etiopathogenesis of HPV-associated oropharyngeal carcinoma and provide targeted therapeutic options. J Otolaryngol Head Neck Surg 2017; 46:52. [PMID: 28818106 PMCID: PMC5561614 DOI: 10.1186/s40463-017-0230-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 08/10/2017] [Indexed: 12/18/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Robert E. Brown
- Department of Pathology and Laboratory Medicine, at UT Health McGovern Medical School, Houston, TX USA
| | - Syed Naqvi
- Department of Otorhinolaryngology, Head and Neck Surgery at UT Health McGovern Medical School, Houston, TX USA
| | - Mary F. McGuire
- Department of Pathology and Laboratory Medicine, at UT Health McGovern Medical School, Houston, TX USA
| | - Jamie Buryanek
- Department of Pathology and Laboratory Medicine, at UT Health McGovern Medical School, Houston, TX USA
| | - Ron J. Karni
- Department of Otorhinolaryngology, Head and Neck Surgery at UT Health McGovern Medical School, Houston, TX USA
| |
Collapse
|
9
|
Xin P, Li C, Zheng Y, Peng Q, Xiao H, Huang Y, Zhu X. Efficacy of the dual PI3K and mTOR inhibitor NVP-BEZ235 in combination with imatinib mesylate against chronic myelogenous leukemia cell lines. Drug Des Devel Ther 2017; 11:1115-1126. [PMID: 28435223 PMCID: PMC5388256 DOI: 10.2147/dddt.s132092] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin (PI3K/Akt/mTOR) pathway is a therapy target of cancer. We aimed to confirm the effect of dual PI3K/mTOR inhibitor NVP-BEZ235 on proliferation, apoptosis, and autophagy of chronic myelogenous leukemia (CML) cells and sensitivity of tyrosine kinase inhibitor in vitro. METHODS Two human CML cell lines, K562 and KBM7R (T315I mutant strain), were used. The proliferation of CML cells was detected by MTS (Owen's reagent) assay. Cell cycle and apoptosis assay were examined by flow cytometric analysis. The phosphorylation levels and the expression levels were both evaluated by Western blot analysis. NVP-BEZ235 in combination with imatinib was also used to reveal the effect on proliferation and apoptosis. RESULTS NVP-BEZ235 significantly inhibited the proliferation in a time- and dose-dependent manner, and the half-maximal inhibitory concentration values of NVP-BEZ235 inhibiting the proliferation of K562 and KBM7R were 0.37±0.21 and 0.43±0.27 μmol/L, respectively, after 48 h. Cell apoptosis assay showed that NVP-BEZ235 significantly increased the late apoptotic cells. Cell cycle analysis indicated that the cells were mostly arrested in G1/G0 phase after treatment by NVP-BEZ235. In addition, results also found that, after treatment by NVP-BEZ235, phosphorylation levels of Akt kinase and S6K kinase significantly reduced, and the expression levels of cleaved caspase-3 significantly increased; meanwhile, the expression levels of caspase-3, B-cell lymphoma-2, cyclin D1, and cyclin D2 significantly decreased, and the ratio of LC3II/LC3I was significantly increased with increased LC3II expression level. Moreover, imatinib in combination with NVP-BEZ235 induced a more pronounced colony growth inhibition than imatinib alone. CONCLUSION NVP-BEZ235 effectively inhibited cell proliferation by G0/G1 cell cycle arrest and induced apoptosis through deregulating PI3K/Akt/mTOR pathway in CML cells; in addition, NVP-BEZ235 can enhance cell autophagy, and is conducive to raising CML cell sensitivity to imatinib to inhibit the growth of imatinib-resistant cells.
Collapse
Affiliation(s)
- Pengliang Xin
- Department of Haematology, First Hospital of Quanzhou Affiliated to Fujian Medical University, Licheng, Quanzhou, Fujian Province, China
| | - Chuntuan Li
- Department of Haematology, First Hospital of Quanzhou Affiliated to Fujian Medical University, Licheng, Quanzhou, Fujian Province, China
| | - Yan Zheng
- Department of Haematology, First Hospital of Quanzhou Affiliated to Fujian Medical University, Licheng, Quanzhou, Fujian Province, China
| | - Qunyi Peng
- Department of Haematology, First Hospital of Quanzhou Affiliated to Fujian Medical University, Licheng, Quanzhou, Fujian Province, China
| | - Huifang Xiao
- Department of Haematology, First Hospital of Quanzhou Affiliated to Fujian Medical University, Licheng, Quanzhou, Fujian Province, China
| | - Yuanling Huang
- Department of Haematology, First Hospital of Quanzhou Affiliated to Fujian Medical University, Licheng, Quanzhou, Fujian Province, China
| | - Xiongpeng Zhu
- Department of Haematology, First Hospital of Quanzhou Affiliated to Fujian Medical University, Licheng, Quanzhou, Fujian Province, China
| |
Collapse
|
10
|
Oh EJ, Kim EK, Yang WI, Yoon SO. Activation of the polycomb repressive complex pathway in the bone marrow resident cells of diffuse large B-cell lymphoma patients. Leuk Lymphoma 2016; 57:1921-32. [PMID: 26757888 DOI: 10.3109/10428194.2015.1121261] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The present study investigated the activation of polycomb repressive complex 2 (PRC2) pathway proteins in the resident cells within the bone marrow hematopoietic microenvironment of diffuse large B-cell lymphoma (DLBCL) patients. PRC2 proteins (enhancer of zeste homolog 2, suppressor of zeste 12 homolog, and embryonic ectoderm development), histone methylation mark (H3K27me3), and c-MYC activation were evaluated in pretreatment bone marrow from 208 DLBLC patients. Positive expression of the PRC2, H3K27me3, and c-MYC in the bone marrow resident cells was more frequent in cases with bone marrow involvement of tumor. The expression among PRC2, H3K27me3 mark, and c-MYC was closely correlated. Positive PRC2 expression in bone marrow resident cells was significantly associated with inferior progression-free survival (PFS) and overall survival (OS) and determined to be an independent prognostic factor of inferior PFS and OS. In conclusion, the PRC pathway was frequently activated in bone marrow resident cells of DLBCL patients, and PRC activation was tumor-related and associated with poor clinical outcomes.
Collapse
Affiliation(s)
- Eun Ji Oh
- a Department of Pathology , Yonsei University College of Medicine , Seoul , Korea
| | - Eun Kyung Kim
- a Department of Pathology , Yonsei University College of Medicine , Seoul , Korea
| | - Woo Ick Yang
- a Department of Pathology , Yonsei University College of Medicine , Seoul , Korea
| | - Sun Och Yoon
- a Department of Pathology , Yonsei University College of Medicine , Seoul , Korea
| |
Collapse
|
11
|
Li C, Xin P, Xiao H, Zheng Y, Huang Y, Zhu X. The dual PI3K/mTOR inhibitor NVP-BEZ235 inhibits proliferation and induces apoptosis of burkitt lymphoma cells. Cancer Cell Int 2015; 15:65. [PMID: 26130968 PMCID: PMC4486138 DOI: 10.1186/s12935-015-0213-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 06/03/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin (PI3K/Akt/mTOR) pathway is a therapy target of cancer. We aimed to confirm the effect of dual PI3K/mTOR inhibitor NVP-BEZ235 on cell proliferation and apoptosis in Burkitt lymphoma (BL) cells. METHODS Two human BL cell lines, CA46 and RAJI were used in this study. The proliferation of BL cells was detected by manganese tricarbonyl transfer (MTT) assay. Cell cycle and apoptosis assay were examined by flow cytometric analysis. The phosphorylation levels of AKT (Thr308), AKT (Ser473), and RPS6 were evaluated by western blot analysis. RESULTS NVP-BEZ235 significantly inhibited the proliferation of BL cells (CA46 and RAJI) and the inhibition effect was time and dose-dependent. Cell cycle analysis indicated that the cells (CA46 and RAJI) were mostly arrested in G1/G0 phase. Cell apoptosis assay showed that the late apoptotic cells were significantly increased after 72 h treatment by 100 nmol/L of NVP-BEZ235. In addition, results also found that NVP-BEZ235 reduced the phosphorylation levels of AKT (Thr308), AKT (Ser473), and PRS6 in BL cells (CA46 and RAJI). Moreover, this inhibition effect on phosphorylation was dose-dependent. CONCLUSIONS NVP-BEZ235 effectively inhibited cell proliferation by G0/G1 cell-cycle arrest and induced apoptosis through deregulating PI3K/Akt/mTOR pathway in BL cells.
Collapse
Affiliation(s)
- Chuntuan Li
- Department of Haematology, First Hospital of Quanzhou Affiliated to Fujian Medical University, 248 East Street, Licheng District, Quanzhou, 362000 Fujian Province China
| | - Pengliang Xin
- Department of Haematology, First Hospital of Quanzhou Affiliated to Fujian Medical University, 248 East Street, Licheng District, Quanzhou, 362000 Fujian Province China
| | - Huifang Xiao
- Department of Haematology, First Hospital of Quanzhou Affiliated to Fujian Medical University, 248 East Street, Licheng District, Quanzhou, 362000 Fujian Province China
| | - Yan Zheng
- Department of Haematology, First Hospital of Quanzhou Affiliated to Fujian Medical University, 248 East Street, Licheng District, Quanzhou, 362000 Fujian Province China
| | - Yuanling Huang
- Department of Haematology, First Hospital of Quanzhou Affiliated to Fujian Medical University, 248 East Street, Licheng District, Quanzhou, 362000 Fujian Province China
| | - Xiongpeng Zhu
- Department of Haematology, First Hospital of Quanzhou Affiliated to Fujian Medical University, 248 East Street, Licheng District, Quanzhou, 362000 Fujian Province China
| |
Collapse
|
12
|
Vélez-Pérez A, Huang RSP, Everett JM, Guo S, Buryanek J, Karni RJ, Brown RE. Carcinosarcoma of the parotid: investigating its biology with morphoproteomics. Int J Surg Pathol 2014; 23:116-21. [PMID: 25515562 DOI: 10.1177/1066896914562278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Carcinosarcoma of the parotid is a rare biphasic malignant neoplasm comprised of both carcinomatous and sarcomatous components representing approximately 0.4% of all malignant salivary gland neoplasms. We report a case of a 55-year-old Caucasian man who presented with a progressively enlarging left facial mass. Histopathological evaluation of the tumoral tissue revealed a high grade, mixed epithelial and mesenchymal malignant tumor, most consistent with a carcinosarcoma of the parotid. Morphoproteomic analysis was performed and revealed expression of secreted protein acidic and rich in cysteine (SPARC); glioma-associated oncogen protein 2 (Gli2); and phosphorylated signal transducer and activator of transcription (p-STAT3 [Tyr705]) in the carcinomatous and malignant mesenchymal components. These aforementioned markers have been linked to the epithelial-mesenchymal transition in which epithelial cells lose their characteristics and phenotypically become mesenchymal cells. This finding allows us to further understand the biology of the 2 cellular components of the carcinosarcoma as having a monoclonal origin.
Collapse
Affiliation(s)
| | - Richard S P Huang
- The University of Texas at Houston Health Science Center, Houston, TX, USA
| | - Jamie M Everett
- The University of Texas at Houston Health Science Center, Houston, TX, USA
| | - Shan Guo
- The University of Texas at Houston Health Science Center, Houston, TX, USA
| | - Jamie Buryanek
- The University of Texas at Houston Health Science Center, Houston, TX, USA
| | - Ron J Karni
- The University of Texas at Houston Health Science Center, Houston, TX, USA
| | - Robert E Brown
- The University of Texas at Houston Health Science Center, Houston, TX, USA
| |
Collapse
|
13
|
Shin D, Arthur G, Popescu M, Korkin D, Shyu CR. Uncovering influence links in molecular knowledge networks to streamline personalized medicine. J Biomed Inform 2014; 52:394-405. [PMID: 25150201 DOI: 10.1016/j.jbi.2014.08.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 08/04/2014] [Accepted: 08/08/2014] [Indexed: 01/10/2023]
Abstract
OBJECTIVES We developed Resource Description Framework (RDF)-induced InfluGrams (RIIG) - an informatics formalism to uncover complex relationships among biomarker proteins and biological pathways using the biomedical knowledge bases. We demonstrate an application of RIIG in morphoproteomics, a theranostic technique aimed at comprehensive analysis of protein circuitries to design effective therapeutic strategies in personalized medicine setting. METHODS RIIG uses an RDF "mashup" knowledge base that integrates publicly available pathway and protein data with ontologies. To mine for RDF-induced Influence Links, RIIG introduces notions of RDF relevancy and RDF collider, which mimic conditional independence and "explaining away" mechanism in probabilistic systems. Using these notions and constraint-based structure learning algorithms, the formalism generates the morphoproteomic diagrams, which we call InfluGrams, for further analysis by experts. RESULTS RIIG was able to recover up to 90% of predefined influence links in a simulated environment using synthetic data and outperformed a naïve Monte Carlo sampling of random links. In clinical cases of Acute Lymphoblastic Leukemia (ALL) and Mesenchymal Chondrosarcoma, a significant level of concordance between the RIIG-generated and expert-built morphoproteomic diagrams was observed. In a clinical case of Squamous Cell Carcinoma, RIIG allowed selection of alternative therapeutic targets, the validity of which was supported by a systematic literature review. We have also illustrated an ability of RIIG to discover novel influence links in the general case of the ALL. CONCLUSIONS Applications of the RIIG formalism demonstrated its potential to uncover patient-specific complex relationships among biological entities to find effective drug targets in a personalized medicine setting. We conclude that RIIG provides an effective means not only to streamline morphoproteomic studies, but also to bridge curated biomedical knowledge and causal reasoning with the clinical data in general.
Collapse
Affiliation(s)
- Dmitriy Shin
- University of Missouri, School of Medicine, Department of Pathology and Anatomical Sciences, Columbia, MO 65212, United States; University of Missouri, Graduate School, MU Informatics Institute, Columbia, MO 65211, United States.
| | - Gerald Arthur
- University of Missouri, School of Medicine, Department of Pathology and Anatomical Sciences, Columbia, MO 65212, United States; University of Missouri, Graduate School, MU Informatics Institute, Columbia, MO 65211, United States
| | - Mihail Popescu
- University of Missouri, School of Medicine, Department of Health Management and Informatics, Columbia, MO 65212, United States; University of Missouri, Graduate School, MU Informatics Institute, Columbia, MO 65211, United States; University of Missouri, College of Engineering, Department of Computer Science, Columbia, MO 65211, United States
| | - Dmitry Korkin
- Worcester Polytechnic Institute, Department of Computer Science, Department of Biology and Biotechnology, Department of Applied Math, Worcester, MA 01609, United States
| | - Chi-Ren Shyu
- University of Missouri, Graduate School, MU Informatics Institute, Columbia, MO 65211, United States; University of Missouri, College of Engineering, Department of Electrical and Computer Engineering, Columbia, MO 65211, United States
| |
Collapse
|
14
|
Dhamne S, Brown RE, Covinsky M, Dhamne C, Eldin K, Tatevian N. Clear cell sarcoma of kidney: morphoproteomic analysis reveals genomic correlates and therapeutic options. Pediatr Dev Pathol 2013; 16:20-7. [PMID: 22256788 DOI: 10.2350/11-01-0968-oa.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
We used the morphoproteomic approach to analyze clear cell sarcoma of kidney (CCSK), a rare pediatric renal tumor, for which the exact pathogenesis and reliable diagnostic markers remain inexplicable. The tumor, currently being treated with chemotherapy and radiation therapy before or after radical nephrectomy, has demonstrated improved survival rates after introduction of doxorubicin. Three cases of CCSK were studied. We attempted to decipher the possible pathological mechanisms involved in CCSK and to explore the therapeutic targets and plausible less-toxic chemotherapeutic agents. We propose that cyclin D1 may be a central molecule in the pathogenesis of CCSK, driven mainly by the sonic hedgehog and the nuclear factor-kappa B pathways and secondarily by the mammalian target of rapamycin complex mTORC2/PI3K/Akt pathway, heat shock protein 90, and possibly phospholipase D1. Inclusion of relatively less toxic but effective therapies in the form of statins, 13-cis retinoic acid, curcumin, and 17-AAG in the combinatorial treatment strategies, which can target the involved subcellular pathways, may be considered.
Collapse
|
15
|
Subbiah V, Brown RE, Buryanek J, Trent J, Ashkenazi A, Herbst R, Kurzrock R. Targeting the apoptotic pathway in chondrosarcoma using recombinant human Apo2L/TRAIL (dulanermin), a dual proapoptotic receptor (DR4/DR5) agonist. Mol Cancer Ther 2012; 11:2541-6. [PMID: 22914439 DOI: 10.1158/1535-7163.mct-12-0358] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Recombinant human Apo2L/TRAIL (dulanermin) is based on the ligand for death receptors (DR4 and DR5), which promotes apoptosis. We report a patient with refractory chondrosarcoma who showed a prolonged response to dulanermin and explore mechanisms of response and resistance. This heavily pretreated patient had progressive metastatic chondrosarcoma to the lung. On dulanermin (8 mg/kg i.v. on days 1-5 in a 21-day cycle), the patient achieved a sustained partial response with only subcentimeter nodules remaining. After 62 months of dulanermin treatment, progressive disease in the lungs was noted, and the patient underwent a resection that confirmed chondrosarcoma. DR4 was detected (immunohistochemistry) in the patient's tumor, which may have enabled the response. However, upregulation of prosurvival proteins, namely, phosphorylated (p)-NF-κBp65 (Ser 536), p-STAT3 (Tyr 705), p-ERK 1/2 (Thr 202/Tyr 204), p-mTOR (Ser 2448), FASN, and Bcl-2, were also detected, which may have provided the underlying mechanisms for acquired dulanermin resistance. The patient was restarted on dulanermin and has continued on this treatment for an additional 16 months since surgery (78 months since initiation of treatment), with his most recent computed tomography (CT) scans showing no evidence of disease.
Collapse
Affiliation(s)
- Vivek Subbiah
- The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 455, Houston, Texas 77030, USA.
| | | | | | | | | | | | | |
Collapse
|
16
|
Wolff JE, Brown RE, Buryanek J, Pfister S, Vats TS, Rytting ME. Preliminary experience with personalized and targeted therapy for pediatric brain tumors. Pediatr Blood Cancer 2012; 59:27-33. [PMID: 22162424 DOI: 10.1002/pbc.23402] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Accepted: 09/26/2011] [Indexed: 11/10/2022]
Abstract
BACKGROUND A new generation of anticancer drugs has reached clinical care in common diseases, but their use in rare diseases such as pediatric brain tumors lags behind since conventional clinical trial design requires larger patient numbers. PROCEDURE We designed individualized treatment protocols for pediatric patients with relapsed brain tumors, based upon the patient's treatment history. In addition, each tumor was analyzed with morphoproteomics using a panel of markers to show treatment targets, resulting in a list of potential novel drugs to be added to chemotherapy. Here, we present the concept and report the experiences of the first patients enrolled in the program. RESULTS Eleven treatment protocols were designed using morphoproteomic information and given to eight patients. The histological diagnoses included: medulloblastoma (n = 3), glioblastoma multiforme (n = 2), atypical teratoid rhabdoid tumor (n = 1), choroid plexus carcinoma (n = 1), and primitive neuroectodermal tumors (n = 1). Tumor markers included p-ERK, Topoisomerase IIa, Bcl-2, VEGF-A, p-STAT3, ER-beta, p-mTOR, and p-NF-kappaBp65. The novel agents included sorafenib, bevacizumab, fulvestrant, rapamycin, bortezomib, and curcumin. The response to the first protocol was complete response: 1, partial response: 1, stable disease: 0, progressive disease: 4, and continuous complete remission: 2. The median Event-Free Survival was 0.32 year ± 0.4. For the comparison with the institutional control group, the individual response probability was calculated. The observed response was superior to the historical controls (P = 0.006 Whitman U-test). CONCLUSION This approach warrants further, systematic evaluation as proof of concept and then expansion to drug-specific hypotheses.
Collapse
Affiliation(s)
- Johannes E Wolff
- Tufts Medical Center, The Floating Hospital for Children, Boston, Massachusetts 02111, USA.
| | | | | | | | | | | |
Collapse
|
17
|
McGuire MF, Sriram Iyengar M, Mercer DW. Data driven linear algebraic methods for analysis of molecular pathways: application to disease progression in shock/trauma. J Biomed Inform 2012; 45:372-87. [PMID: 22200681 PMCID: PMC3346262 DOI: 10.1016/j.jbi.2011.12.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Revised: 12/09/2011] [Accepted: 12/10/2011] [Indexed: 12/24/2022]
Abstract
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.
Collapse
Affiliation(s)
- Mary F McGuire
- Department of Pathology and Laboratory Medicine, Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA.
| | | | | |
Collapse
|
18
|
Liu J, Brown RE. Morphoproteomics demonstrates activation of mammalian target of rapamycin pathway in papillary thyroid carcinomas with nuclear translocation of MTOR in aggressive histological variants. Mod Pathol 2011; 24:1553-9. [PMID: 21822208 DOI: 10.1038/modpathol.2011.121] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
We used morphoproteomics to investigate mammalian target of rapamycin (MTOR) signaling pathway in papillary thyroid carcinomas and correlated the results with clinicopathological parameters. Archival paraffin-embedded tissue of papillary thyroid carcinomas was obtained from 30 patients, including 15 classical type and 8 follicular, 4 tall-cell, 1 columnar-cell, 1 diffuse sclerosing and 1 cribriform variants. Immunohistochemical stains were performed for three phosphorylated (p) protein analytes: p-MTOR (Ser2448), p-Akt (Ser473) and p-p70S6K (Thr389). Chromogenic signals and subcellular compartmentalization (nuclear, cytoplasmic and plasmalemmal) were evaluated. Clinicopathological parameters were reviewed. Immunoreactivities for p-MTOR, p-Akt and p-p70S6K were observed in all papillary thyroid carcinomas. In addition to an expression of p-MTOR in cytoplasmic location, nuclear translocation of p-MTOR with variable loss of plasmalemmal expression, and with concomitant nuclear expression of p-Akt, was also identified in all tall-cell, columnar-cell and diffuse sclerosing variants of papillary thyroid carcinoma. There were no significant differences in the clinicopathological parameters, including tumor size, extrathyroidal extension, angioinvasion and nodal metastases between the groups with and without nuclear expression of p-MTOR (P>0.05). The expressions of p-MTOR in cytoplasmic and/or plasmalemmal locations with the concomitant immunoreactivity for p-p70S6K in all papillary thyroid carcinomas indicate the activation of MTOR complex 1 pathway. The nuclear translocation of p-MTOR evidences the activation of MTOR complex 2 and is identified only in the known aggressive histological variants of papillary thyroid carcinoma, including tall-cell, columnar-cell and diffuse sclerosing variants. Thus, these results suggest the constitutive activation of MTOR signaling pathway in papillary thyroid carcinomas and provide a new insight of biogenetic basis for the aggressive histological variants of papillary thyroid carcinoma. The pattern of expression of p-MTOR in papillary thyroid carcinomas may serve as a diagnostic/prognostic marker and a potential therapeutic target.
Collapse
Affiliation(s)
- Jing Liu
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at Houston Medical School, Houston, TX, USA.
| | | |
Collapse
|
19
|
Subbiah V, Naing A, Brown RE, Chen H, Doyle L, LoRusso P, Benjamin R, Anderson P, Kurzrock R. Targeted morphoproteomic profiling of Ewing's sarcoma treated with insulin-like growth factor 1 receptor (IGF1R) inhibitors: response/resistance signatures. PLoS One 2011; 6:e18424. [PMID: 21494688 PMCID: PMC3071831 DOI: 10.1371/journal.pone.0018424] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2010] [Accepted: 03/05/2011] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Insulin-like growth factor 1 receptor (IGF1R) targeted therapies have resulted in responses in a small number of patients with advanced metastatic Ewing's sarcoma. We performed morphoproteomic profiling to better understand response/resistance mechanisms of Ewing's sarcoma to IGF1R inhibitor-based therapy. METHODOLOGY/PRINCIPAL FINDINGS This pilot study assessed two patients with advanced Ewing's sarcoma treated with IGF1R antibody alone followed by combined IGF1R inhibitor plus mammalian target of rapamycin (mTOR) inhibitor treatment once resistance to single-agent IGF1R inhibitor developed. Immunohistochemical probes were applied to detect p-mTOR (Ser2448), p-Akt (Ser473), p-ERK1/2 (Thr202/Tyr204), nestin, and p-STAT3 (Tyr 705) in the original and recurrent tumor. The initial remarkable radiographic responses to IGF1R-antibody therapy was followed by resistance and then response to combined IGF1R plus mTOR inhibitor therapy in both patients, and then resistance to the combination regimen in one patient. In patient 1, upregulation of p-Akt and p-mTOR in the tumor that relapsed after initial response to IGF1R antibody might explain the resistance that developed, and the subsequent response to combined IGF1R plus mTOR inhibitor therapy. In patient 2, upregulation of mTOR was seen in the primary tumor, perhaps explaining the initial response to the IGF1R and mTOR inhibitor combination, while the resistant tumor that emerged showed activation of the ERK pathway as well. CONCLUSION/SIGNIFICANCE Morphoproteomic analysis revealed that the mTOR pathway was activated in these two patients with advanced Ewing's sarcoma who showed response to combined IGF1R and mTOR inhibition, and the ERK pathway in the patient in whom resistance to this combination emerged. Our pilot results suggests that morphoproteomic assessment of signaling pathway activation in Ewing's sarcoma merits further investigation as a guide to understanding response and resistance signatures.
Collapse
Affiliation(s)
- Vivek Subbiah
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Streckfus CF, Brown RE, Bull JM. Proteomics, morphoproteomics, saliva and breast cancer: An emerging approach to guide the delivery of individualised thermal therapy, thermochemotherapy and monitor therapy response. Int J Hyperthermia 2010; 26:649-61. [DOI: 10.3109/02656736.2010.506470] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
21
|
Niederberger E, Geisslinger G. Analysis of NF-kappaB signaling pathways by proteomic approaches. Expert Rev Proteomics 2010; 7:189-203. [PMID: 20377387 DOI: 10.1586/epr.10.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
NF-kappaB is a transcription factor that plays important roles in the regulation of apoptosis and inflammation as well as innate and adaptive immunity. Consequently, dysregulations in the NF-kappaB activation cascade have been associated with the pathogenesis of several diseases such as cancer, atherosclerosis and rheumatoid arthritis. Although NF-kappaB signaling pathways have been extensively investigated in this context, its varying components and targets are far from being completely elucidated. There is still an urgent need for the detection of novel NF-kappaB target proteins, novel interaction partners and novel regulators in the activation cascade, in particular with regard to its role in the aforementioned diseases. Therefore, several groups have performed different proteomic approaches to further investigate NF-kappaB signal transduction pathways. Most of these studies have been carried out in the area of cancer research; however, there are also several analyses in the field of inflammatory or autoimmune diseases. Furthermore, there have been a number of basic investigations that principally examined binding partners or so far unknown target proteins of NF-kappaB-related proteins. With these approaches, a number of novel and interesting proteins have been found that interfere with NF-kappaB signal transduction and might have an impact on NF-kappaB-related diseases. The results of these studies are summarized and discussed in this review.
Collapse
Affiliation(s)
- Ellen Niederberger
- Pharmazentrum Frankfurt/ZAFES, Institut für Klinische Pharmakologie, Klinikum der Goethe-Universität Frankfurt, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany.
| | | |
Collapse
|
22
|
Hernandez JS, Dale JC, Bennet KE, Varkey P. Challenges and opportunities for medical directors in pathology and laboratory medicine: standardization, integration, and innovation. Am J Clin Pathol 2010; 133:8-13. [PMID: 20023253 DOI: 10.1309/ajcpsb4al5ehawtc] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Because the practice of pathology and laboratory medicine evolves rapidly, laboratory medical directors must constantly introduce new tests and services and continue to provide consistent, reliable results for existing tests. Innovations in laboratory medicine are frequently introduced, and the number of commercial vendors of test kits and reagents increases yearly. These innovations, however, may pose barriers to standardization and integration of laboratories and to interpretation of results generated by different laboratories. We propose a practical framework for medical directors to address the seemingly contradictory challenges of standardizing and integrating while simultaneously providing the flexibility to introduce innovations. We recommend initiating standardization first, then integration, while maintaining flexibility for innovation. As organizations strive to create effective processes to enhance value, the role of the laboratory medical director will become critical in resolving the natural tension between standardization/integration and innovation in laboratory medicine and pathology.
Collapse
|
23
|
Brown RE. Morphogenomics and morphoproteomics: a role for anatomic pathology in personalized medicine. Arch Pathol Lab Med 2009; 133:568-79. [PMID: 19391654 DOI: 10.5858/133.4.568] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2008] [Indexed: 11/06/2022]
Affiliation(s)
- Robert E Brown
- Department of Pathology, University of Texas Health Science Center, Houston Medical School, Houston, TX 77030, USA.
| |
Collapse
|
24
|
Zander DS. Transbronchial Fine-Needle Aspiration, Epidermal Growth Factor Receptor Mutations, and the Scorpion's Touch. Chest 2007; 131:1619-20. [PMID: 17565010 DOI: 10.1378/chest.07-0386] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
25
|
Ward K. Microarray technology in obstetrics and gynecology: a guide for clinicians. Am J Obstet Gynecol 2006; 195:364-72. [PMID: 16615920 PMCID: PMC7093878 DOI: 10.1016/j.ajog.2005.12.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2005] [Revised: 11/29/2005] [Accepted: 12/05/2005] [Indexed: 11/28/2022]
Abstract
Microarrays can be constructed with dozens to millions of probes on their surface to allow high-throughput analyses of many biologic processes to be performed simultaneously on the same sample. Microarrays are now widely used for gene expression analysis, deoxyribonucleic acid resequencing, single-nucleotide polymorphism genotyping, and comparative genomic hybridization. Microarray technology is accelerating research in many fields and now microarrays are moving into clinical application. This review discusses the emerging role of microarrays in molecular diagnostics, pathogen detection, oncology, and pharmacogenomics.
Collapse
Affiliation(s)
- Kenneth Ward
- Department of Obstetrics and Gynecology and Women's Health and the Pacific Research Center for Early Human Development, University of Hawaii, John A. Burns School of Medicine, Honolulu, HI 96826, USA.
| |
Collapse
|
26
|
Mundinger GS, Espina V, Liotta LA, Petricoin EF, Calvo KR. Clinical phosphoproteomic profiling for personalized targeted medicine using reverse phase protein microarray. Target Oncol 2006. [DOI: 10.1007/s11523-006-0025-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
27
|
Abstract
CONTEXT The field of molecular pathology is expanding in complexity. To achieve competency, vigilance is required. OBJECTIVE To review the advances in clinically useful molecular biologic techniques and to identify their applications in clinical practice, as presented at the 13th Annual William Beaumont Hospital DNA Symposium. DATA SOURCES The 4 manuscripts submitted were reviewed and their major findings were compared with the literature on the same or related topics. STUDY SELECTION Manuscripts address the use of molecular or immunophenotyping by flow cytometry to evaluate the origin or presence of sepsis, respectively; the use of imatinib mesylate to treat chronic myeloid leukemia and the nature of resistance to imatinib; and the use of 9 and 10 fluorochromes during clinical flow cytometric studies. DATA SYNTHESIS The epidemiologic evaluation of a septic outbreak may be monitored using molecular techniques that track the relatedness of isolates. A potential biomarker for the presence of early sepsis is CD64. Intracellular signal transduction pathways are altered in malignancy. Imatinib mesylate inhibits the BCR-ABL kinase created by translocation of the long arms of chromosomes 9 and 22 in chronic myeloid leukemia. Resistance to imatinib may be secondary to mutation in the BCR-ABL kinase domain or residual leukemic stem cells that imatinib does not kill. The use of 9 or 10 fluorochromes simultaneously during flow cytometry has many clinical advantages; however, software for data analysis is needed. CONCLUSION The current postgenomic era will continue to emphasize the use of microarrays and database software for genomic, transcriptomic, proteomic, nutrigenomic, and pharmacogenomics screening to search for a useful clinical assay. The number of molecular pathologic techniques will expand as additional disease-associated mutations are defined.
Collapse
Affiliation(s)
- Frederick L Kiechle
- Department of Clinical Pathology, William Beaumont Hospital, Royal Oak, MI 48073, USA
| | | | | |
Collapse
|