204 results on '"Sharpless NE"'
Search Results
2. Abstract P1-05-20: Comparing the frequency and types of genetic aberrations between older and younger women with metastatic breast cancer at the University of North Carolina at Chapel Hill
- Author
-
Jolly, TA, primary, Grilley-Olson, JE, additional, Deal, AM, additional, Ivanova, A, additional, Hayward, MC, additional, Benbow, JM, additional, Parker, JS, additional, Patel, NM, additional, Eberhard, DA, additional, Weck, KE, additional, Mieczkowski, P, additional, Dees, EC, additional, Muss, HD, additional, Reeder-Hayes, KE, additional, Earp, HS, additional, Sharpless, NE, additional, Carey, LA, additional, Hayes, DN, additional, and Anders, CK, additional
- Published
- 2017
- Full Text
- View/download PDF
3. Population Pharmacokinetics Modeling of Unbound Efavirenz, Atazanavir, and Ritonavir in HIV‐Infected Subjects With Aging Biomarkers
- Author
-
Dumond, JB, primary, Chen, J, additional, Cottrell, M, additional, Trezza, CR, additional, Prince, HMA, additional, Sykes, C, additional, Torrice, C, additional, White, N, additional, Malone, S, additional, Wang, R, additional, Patterson, KB, additional, Sharpless, NE, additional, and Forrest, A, additional
- Published
- 2016
- Full Text
- View/download PDF
4. p16 INK4a, a Senescence Marker, Influences Tenofovir/Emtricitabine Metabolite Disposition in HIV‐Infected Subjects
- Author
-
Dumond, JB, primary, Collins, JW, additional, Cottrell, ML, additional, Trezza, CR, additional, Prince, HMA, additional, Sykes, C, additional, Torrice, C, additional, White, N, additional, Malone, S, additional, Wang, R, additional, Patterson, KB, additional, Sharpless, NE, additional, and Forrest, A, additional
- Published
- 2016
- Full Text
- View/download PDF
5. IL2 Inducible T-cell Kinase, a Novel Therapeutic Target in Melanoma
- Author
-
Carson, CC, Moschos, SJ, Edmiston, SN, Darr, DB, Nikolaishvili-Feinberg, N, Groben, PA, Zhou, X, Kuan, PF, Pandey, S, Chan, KT, Jordan, JL, Hao, H, Frank, JS, Hopkinson, DA, Gibbs, DC, Alldredge, VD, Parrish, E, Hanna, SC, Berkowitz, P, Rubenstein, DS, Miller, CR, Bear, JE, Ollila, DW, Sharpless, NE, Conway, K, Thomas, NE, Carson, CC, Moschos, SJ, Edmiston, SN, Darr, DB, Nikolaishvili-Feinberg, N, Groben, PA, Zhou, X, Kuan, PF, Pandey, S, Chan, KT, Jordan, JL, Hao, H, Frank, JS, Hopkinson, DA, Gibbs, DC, Alldredge, VD, Parrish, E, Hanna, SC, Berkowitz, P, Rubenstein, DS, Miller, CR, Bear, JE, Ollila, DW, Sharpless, NE, Conway, K, and Thomas, NE
- Abstract
PURPOSE: IL2 inducible T-cell kinase (ITK) promoter CpG sites are hypomethylated in melanomas compared with nevi. The expression of ITK in melanomas, however, has not been established and requires elucidation. EXPERIMENTAL DESIGN: An ITK-specific monoclonal antibody was used to probe sections from deidentified, formalin-fixed paraffin-embedded tumor blocks or cell line arrays and ITK was visualized by IHC. Levels of ITK protein differed among melanoma cell lines and representative lines were transduced with four different lentiviral constructs that each contained an shRNA designed to knockdown ITK mRNA levels. The effects of the selective ITK inhibitor BI 10N on cell lines and mouse models were also determined. RESULTS: ITK protein expression increased with nevus to metastatic melanoma progression. In melanoma cell lines, genetic or pharmacologic inhibition of ITK decreased proliferation and migration and increased the percentage of cells in the G0-G1 phase. Treatment of melanoma-bearing mice with BI 10N reduced growth of ITK-expressing xenografts or established autochthonous (Tyr-Cre/Pten(null)/Braf(V600E)) melanomas. CONCLUSIONS: We conclude that ITK, formerly considered an immune cell-specific protein, is aberrantly expressed in melanoma and promotes tumor development and progression. Our finding that ITK is aberrantly expressed in most metastatic melanomas suggests that inhibitors of ITK may be efficacious for melanoma treatment. The efficacy of a small-molecule ITK inhibitor in the Tyr-Cre/Pten(null)/Braf(V600E) mouse melanoma model supports this possibility.
- Published
- 2015
6. LKB1 loss in melanoma disrupts directional migration toward extracellular matrix cues
- Author
-
Chan, KT, Asokan, SB, King, SJ, Bo, T, Dubose, ES, Liu, W, Berginski, ME, Simon, JM, Davis, IJ, Gomez, SM, Sharpless, NE, Bear, JE, Chan, KT, Asokan, SB, King, SJ, Bo, T, Dubose, ES, Liu, W, Berginski, ME, Simon, JM, Davis, IJ, Gomez, SM, Sharpless, NE, and Bear, JE
- Abstract
Somatic inactivation of the serine/threonine kinase gene STK11/LKB1/PAR-4 occurs in a variety of cancers, including ∼10% of melanoma. However, how the loss of LKB1 activity facilitates melanoma invasion and metastasis remains poorly understood. In LKB1-null cells derived from an autochthonous murine model of melanoma with activated Kras and Lkb1 loss and matched reconstituted controls, we have investigated the mechanism by which LKB1 loss increases melanoma invasive motility. Using a microfluidic gradient chamber system and time-lapse microscopy, in this paper, we uncover a new function for LKB1 as a directional migration sensor of gradients of extracellular matrix (haptotaxis) but not soluble growth factor cues (chemotaxis). Systematic perturbation of known LKB1 effectors demonstrated that this response does not require canonical adenosine monophosphate-activated protein kinase (AMPK) activity but instead requires the activity of the AMPK-related microtubule affinity-regulating kinase (MARK)/PAR-1 family kinases. Inhibition of the LKB1-MARK pathway facilitated invasive motility, suggesting that loss of the ability to sense inhibitory matrix cues may promote melanoma invasion.
- Published
- 2014
7. p16INK4a, a Senescence Marker, Influences Tenofovir/Emtricitabine Metabolite Disposition in HIV-Infected Subjects.
- Author
-
Dumond, JB, Collins, JW, Cottrell, ML, Trezza, CR, Prince, HMA, Sykes, C, Torrice, C, White, N, Malone, S, Wang, R, Patterson, KB, Sharpless, NE, and Forrest, A
- Subjects
OLD age ,HIV-positive persons ,TENOFOVIR ,EMTRICITABINE ,GENE expression ,METABOLITES ,NUCLEOSIDES ,PHOSPHORYLATION - Abstract
The goal of this study was to explore the relationships between tenofovir (TFV) and emtricitabine (FTC) disposition and markers of biologic aging, such as the frailty phenotype and p16
INK4a gene expression. Chronologic age is often explored in population pharmacokinetic (PK) analyses, and can be uninformative in capturing the impact of aging on physiology, particularly in human immunodeficiency virus (HIV)-infected patients. Ninety-one HIV-infected participants provided samples to quantify plasma concentrations of TFV/FTC, as well as peripheral blood mononuclear cell (PBMC) samples for intracellular metabolite concentrations; 12 participants provided 11 samples, and 79 participants provided 4 samples, over a dosing interval. Nonlinear mixed effects modeling of TFV/FTC and their metabolites suggests a relationship between TFV/FTC metabolite clearance (CL) from PBMCs and the expression of p16INK4a , a marker of cellular senescence. This novel approach to quantifying the influence of aging on PKs provides rationale for further work investigating the relationships between senescence and nucleoside phosphorylation and transport. [ABSTRACT FROM AUTHOR]- Published
- 2017
- Full Text
- View/download PDF
8. Population Pharmacokinetics Modeling of Unbound Efavirenz, Atazanavir, and Ritonavir in HIV-Infected Subjects With Aging Biomarkers.
- Author
-
Dumond, JB, Chen, J, Cottrell, M, Trezza, CR, Prince, HMA, Sykes, C, Torrice, C, White, N, Malone, S, Wang, R, Patterson, KB, Sharpless, NE, and Forrest, A
- Subjects
PHARMACOKINETICS ,AGING ,EFAVIRENZ ,HIV ,RITONAVIR ,ATAZANAVIR ,BODY mass index - Abstract
Unbound drug is the pharmacodynamically relevant concentration. This study aimed to determine if chronologic age or markers of biologic aging, such as the frailty phenotype and p16
INK4a gene expression, altered unbound pharmacokinetics (PKs) of efavirenz (EFV) and atazanavir/ritonavir (ATV/RTV). Sixty human immunodeficiency virus (HIV)-infected participants receiving EFV and 31 receiving ATV/RTV provided 1 to 11 samples to quantify total and unbound plasma concentrations. Population PK models with total and unbound concentrations simultaneously described are developed for each drug. The unbound fractions for EFV, ATV, and RTV are 0.65%, 5.67%, and 0.63%, respectively. Covariate analysis suggests RTV unbound PK is sensitive to body size; unbound fraction of RTV is 34% lower with body mass index (BMI) above 30 kg/m2 . No alterations in drug clearance or unbound fraction with age, frailty, or p16INK4a expression were observed. Assessing functional and physiologic aging markers to inform potential PK changes is necessary to determine if drug/dosing changes are warranted in the aging population. [ABSTRACT FROM AUTHOR]- Published
- 2017
- Full Text
- View/download PDF
9. Intravital imaging of a spheroid-based orthotopic model of melanoma in the mouse ear skin
- Author
-
Chan, KT, Jones, SW, Brighton, HE, Bo, T, Cochran, SD, Sharpless, NE, Bear, JE, Chan, KT, Jones, SW, Brighton, HE, Bo, T, Cochran, SD, Sharpless, NE, and Bear, JE
- Abstract
Multiphoton microscopy is a powerful tool that enables the visualization of fluorescently tagged tumor cells and their stromal interactions within tissues in vivo. We have developed an orthotopic model of implanting multicellular melanoma tumor spheroids into the dermis of the mouse ear skin without the requirement for invasive surgery. Here, we demonstrate the utility of this approach to observe the primary tumor, single cell actin dynamics, and tumor-associated vasculature. These methods can be broadly applied to investigate an array of biological questions regarding tumor cell behavior in vivo.
- Published
- 2013
10. LKB1/STK11 Inactivation Leads to Expansion of a Prometastatic Tumor Subpopulation in Melanoma
- Author
-
Liu, W, Monahan, KB, Pfefferle, AD, Shimamura, T, Sorrentino, J, Chan, KT, Roadcap, DW, Ollila, DW, Thomas, NE, Castrillon, DH, Miller, CR, Perou, CM, Wong, K-K, Bear, JE, Sharpless, NE, Liu, W, Monahan, KB, Pfefferle, AD, Shimamura, T, Sorrentino, J, Chan, KT, Roadcap, DW, Ollila, DW, Thomas, NE, Castrillon, DH, Miller, CR, Perou, CM, Wong, K-K, Bear, JE, and Sharpless, NE
- Abstract
Germline mutations in LKB1 (STK11) are associated with the Peutz-Jeghers syndrome (PJS), which includes aberrant mucocutaneous pigmentation, and somatic LKB1 mutations occur in 10% of cutaneous melanoma. By somatically inactivating Lkb1 with K-Ras activation (±p53 loss) in murine melanocytes, we observed variably pigmented and highly metastatic melanoma with 100% penetrance. LKB1 deficiency resulted in increased phosphorylation of the SRC family kinase (SFK) YES, increased expression of WNT target genes, and expansion of a CD24(+) cell population, which showed increased metastatic behavior in vitro and in vivo relative to isogenic CD24(-) cells. These results suggest that LKB1 inactivation in the context of RAS activation facilitates metastasis by inducing an SFK-dependent expansion of a prometastatic, CD24(+) tumor subpopulation.
- Published
- 2012
11. Genetic "lnc"-age of noncoding RNAs to human disease.
- Author
-
Troy A, Sharpless NE, Troy, Andrew, and Sharpless, Norman E
- Abstract
The list of functions of long noncoding RNAs (lncRNAs) in human tissues is rapidly growing. To further underscore their critical role in human health, two reports in this issue of JCI associate altered expression of novel lncRNAs with the heritable syndromes HELLP and brachydactyly type E. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
12. PARP and cancer--if it's broke, don't fix it.
- Author
-
Carey LA, Sharpless NE, Carey, Lisa A, and Sharpless, Norman E
- Published
- 2011
- Full Text
- View/download PDF
13. A phase II study of troglitazone, an activator of the PPARgamma receptor, in patients with chemotherapy-resistant metastatic colorectal cancer.
- Author
-
Kulke MH, Demetri GD, Sharpless NE, Ryan DP, Shivdasani R, Clark JS, Spiegelman BM, Kim H, Mayer RJ, Fuchs CS, Kulke, Matthew H, Demetri, George D, Sharpless, Norman E, Ryan, David P, Shivdasani, Ramesh, Clark, Jeffrey S, Spiegelman, Bruce M, Kim, Haesook, Mayer, Robert J, and Fuchs, Charles S
- Abstract
Purpose: Troglitazone, a potent activator of the peroxisome proliferator-activated receptor-gamma, induces tumor differentiation in human liposarcomas and causes regression of tumors that are derived from human colon cancer cells in nude mice. We therefore assessed the efficacy of troglitazone in the treatment of metastatic colon cancer in humans.Methods: Twenty-five patients with metastatic colorectal cancer were treated with oral troglitazone. Patients were followed up for evidence of toxicity, tumor response, and survival.Results: The treatment was well tolerated: no grade 3/4 treatment-related toxicities were observed. However, no objective tumor responses were noted, and all 25 patients had progressive disease as their best response to therapy. The median progression-free survival time was only 1.6 months, and the median survival time was 3.9 months.Discussion: Troglitazone is not an active agent for the treatment of metastatic colorectal cancer. [ABSTRACT FROM AUTHOR]- Published
- 2002
- Full Text
- View/download PDF
14. Bridging the Gap in Cancer Clinical Trial Funding.
- Author
-
Abdou Y and Sharpless NE
- Published
- 2024
- Full Text
- View/download PDF
15. Expression of Concern: Exploiting Drug Addiction Mechanisms to Select against MAPKi-Resistant Melanoma.
- Author
-
Hong A, Moriceau G, Sun L, Lomeli S, Piva M, Damoiseaux R, Holmen SL, Sharpless NE, Hugo W, and Lo RS
- Published
- 2024
- Full Text
- View/download PDF
16. Author Correction: An oncogenic Ezh2 mutation induces tumors through global redistribution of histone 3 lysine 27 trimethylation.
- Author
-
Souroullas GP, Jeck WR, Parker JS, Simon JM, Liu JY, Paulk J, Xiong J, Clark KS, Fedoriw Y, Qi J, Burd CE, Bradner JE, and Sharpless NE
- Published
- 2024
- Full Text
- View/download PDF
17. The Childhood Cancer Data Initiative: Using the Power of Data to Learn From and Improve Outcomes for Every Child and Young Adult With Pediatric Cancer.
- Author
-
Flores-Toro JA, Jagu S, Armstrong GT, Arons DF, Aune GJ, Chanock SJ, Hawkins DS, Heath A, Helman LJ, Janeway KA, Levine JE, Miller E, Penberthy L, Roberts CWM, Shalley ER, Shern JF, Smith MA, Staudt LM, Volchenboum SL, Zhang J, Zenklusen JC, Lowy DR, Sharpless NE, Guidry Auvil JM, Kerlavage AR, Widemann BC, Reaman GH, Kibbe WA, and Doroshow JH
- Subjects
- Adolescent, United States epidemiology, Humans, Child, Young Adult, Ecosystem, Data Collection, National Cancer Institute (U.S.), Neoplasms therapy
- Abstract
Data-driven basic, translational, and clinical research has resulted in improved outcomes for children, adolescents, and young adults (AYAs) with pediatric cancers. However, challenges in sharing data between institutions, particularly in research, prevent addressing substantial unmet needs in children and AYA patients diagnosed with certain pediatric cancers. Systematically collecting and sharing data from every child and AYA can enable greater understanding of pediatric cancers, improve survivorship, and accelerate development of new and more effective therapies. To accomplish this goal, the Childhood Cancer Data Initiative (CCDI) was launched in 2019 at the National Cancer Institute. CCDI is a collaborative community endeavor supported by a 10-year, $50-million (in US dollars) annual federal investment. CCDI aims to learn from every patient diagnosed with a pediatric cancer by designing and building a data ecosystem that facilitates data collection, sharing, and analysis for researchers, clinicians, and patients across the cancer community. For example, CCDI's Molecular Characterization Initiative provides comprehensive clinical molecular characterization for children and AYAs with newly diagnosed cancers. Through these efforts, the CCDI strives to provide clinical benefit to patients and improvements in diagnosis and care through data-focused research support and to build expandable, sustainable data resources and workflows to advance research well past the planned 10 years of the initiative. Importantly, if CCDI demonstrates the success of this model for pediatric cancers, similar approaches can be applied to adults, transforming both clinical research and treatment to improve outcomes for all patients with cancer.
- Published
- 2023
- Full Text
- View/download PDF
18. Topical therapy for regression and melanoma prevention of congenital giant nevi.
- Author
-
Choi YS, Erlich TH, von Franque M, Rachmin I, Flesher JL, Schiferle EB, Zhang Y, Pereira da Silva M, Jiang A, Dobry AS, Su M, Germana S, Lacher S, Freund O, Feder E, Cortez JL, Ryu S, Babila Propp T, Samuels YL, Zakka LR, Azin M, Burd CE, Sharpless NE, Liu XS, Meyer C, Austen WG Jr, Bojovic B, Cetrulo CL Jr, Mihm MC, Hoon DS, Demehri S, Hawryluk EB, and Fisher DE
- Subjects
- Animals, Heterografts, Humans, Mice, Neoplasm Transplantation, Melanoma drug therapy, Melanoma pathology, Nevus, Pigmented congenital, Nevus, Pigmented drug therapy, Nevus, Pigmented pathology, Skin Neoplasms drug therapy, Skin Neoplasms pathology, Skin Neoplasms prevention & control
- Abstract
Giant congenital melanocytic nevi are NRAS-driven proliferations that may cover up to 80% of the body surface. Their most dangerous consequence is progression to melanoma. This risk often triggers preemptive extensive surgical excisions in childhood, producing severe lifelong challenges. We have presented preclinical models, including multiple genetically engineered mice and xenografted human lesions, which enabled testing locally applied pharmacologic agents to avoid surgery. The murine models permitted the identification of proliferative versus senescent nevus phases and treatments targeting both. These nevi recapitulated the histologic and molecular features of human giant congenital nevi, including the risk of melanoma transformation. Cutaneously delivered MEK, PI3K, and c-KIT inhibitors or proinflammatory squaric acid dibutylester (SADBE) achieved major regressions. SADBE triggered innate immunity that ablated detectable nevocytes, fully prevented melanoma, and regressed human giant nevus xenografts. These findings reveal nevus mechanistic vulnerabilities and suggest opportunities for topical interventions that may alter the therapeutic options for children with congenital giant nevi., Competing Interests: Declaration of interests D.E.F. has a financial interest in Soltego, a company developing salt inducible kinase inhibitors for topical skin-darkening treatments that might be used for a broad set of human applications. The interests of D.E.F. were reviewed and are managed by Massachusetts General Hospital and Partners HealthCare in accordance with their conflict-of-interest policies. C.L.C. has a financial interest in 4Immune, a company developing cell therapy treatments that can be used for a broad set of human applications. The interests of C.L.C were reviewed and are managed by Mass General Brigham in accordance with their conflict-of-interest policies. X.S.L. is a cofounder, board member, SAB member, and consultant of GV20 Oncotherapy and its subsidiaries; stockholder of BMY, TMO, WBA, ABT, ABBV, and JNJ; and received research funding from Takeda, Sanofi, Bristol Myers Squibb, and Novartis. M.C.M. discloses consulting relationship with Novartis, Advisory Board with BioCoz and Caliber ID, and author royalties with Wiley & Sons., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
19. Antibodies to Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) in All of Us Research Program Participants, 2 January to 18 March 2020.
- Author
-
Althoff KN, Schlueter DJ, Anton-Culver H, Cherry J, Denny JC, Thomsen I, Karlson EW, Havers FP, Cicek MS, Thibodeau SN, Pinto LA, Lowy D, Malin BA, Ohno-Machado L, Williams C, Goldstein D, Kouame A, Ramirez A, Roman A, Sharpless NE, Gebo KA, and Schully SD
- Subjects
- Antibodies, Viral, Enzyme-Linked Immunosorbent Assay, Humans, Immunoglobulin G, SARS-CoV-2, Sensitivity and Specificity, COVID-19 diagnosis, Population Health
- Abstract
Background: With limited severe acute respiratory syndrome coronavirus (SARS-CoV-2) testing capacity in the United States at the start of the epidemic (January-March 2020), testing was focused on symptomatic patients with a travel history throughout February, obscuring the picture of SARS-CoV-2 seeding and community transmission. We sought to identify individuals with SARS-CoV-2 antibodies in the early weeks of the US epidemic., Methods: All of Us study participants in all 50 US states provided blood specimens during study visits from 2 January to 18 March 2020. Participants were considered seropositive if they tested positive for SARS-CoV-2 immunoglobulin G (IgG) antibodies with the Abbott Architect SARS-CoV-2 IgG enzyme-linked immunosorbent assay (ELISA) and the EUROIMMUN SARS-CoV-2 ELISA in a sequential testing algorithm. The sensitivity and specificity of these ELISAs and the net sensitivity and specificity of the sequential testing algorithm were estimated, along with 95% confidence intervals (CIs)., Results: The estimated sensitivities of the Abbott and EUROIMMUN assays were 100% (107 of 107 [95% CI: 96.6%-100%]) and 90.7% (97 of 107 [83.5%-95.4%]), respectively, and the estimated specificities were 99.5% (995 of 1000 [98.8%-99.8%]) and 99.7% (997 of 1000 [99.1%-99.9%]), respectively. The net sensitivity and specificity of our sequential testing algorithm were 90.7% (97 of 107 [95% CI: 83.5%-95.4%]) and 100.0% (1000 of 1000 [99.6%-100%]), respectively. Of the 24 079 study participants with blood specimens from 2 January to 18 March 2020, 9 were seropositive, 7 before the first confirmed case in the states of Illinois, Massachusetts, Wisconsin, Pennsylvania, and Mississippi., Conclusions: Our findings identified SARS-CoV-2 infections weeks before the first recognized cases in 5 US states., (Published by Oxford University Press for the Infectious Diseases Society of America 2021.)
- Published
- 2022
- Full Text
- View/download PDF
20. A Trans-Governmental Collaboration to Independently Evaluate SARS-CoV-2 Serology Assays.
- Author
-
Pinto LA, Shawar RM, O'Leary B, Kemp TJ, Cherry J, Thornburg N, Miller CN, Gallagher PS, Stenzel T, Schuck B, Owen SM, Kondratovich M, Satheshkumar PS, Schuh A, Lester S, Cassetti MC, Sharpless NE, Gitterman S, and Lowy DR
- Subjects
- COVID-19 diagnosis, COVID-19 epidemiology, COVID-19 virology, Diagnostic Test Approval, Humans, Laboratories, Pandemics, SARS-CoV-2 genetics, Sensitivity and Specificity, Spike Glycoprotein, Coronavirus analysis, Spike Glycoprotein, Coronavirus immunology, United States epidemiology, United States Food and Drug Administration, Antibodies, Viral blood, COVID-19 blood, COVID-19 Serological Testing methods, Enzyme-Linked Immunosorbent Assay methods, SARS-CoV-2 immunology
- Abstract
The emergence of SARS-CoV-2 created a crucial need for serology assays to detect anti-SARS-CoV-2 antibodies, which led to many serology assays entering the market. A trans-government collaboration was created in April 2020 to independently evaluate the performance of commercial SARS-CoV-2 serology assays and help inform U.S. Food and Drug Administration (FDA) regulatory decisions. To assess assay performance, three evaluation panels with similar antibody titer distributions were assembled. Each panel consisted of 110 samples with positive ( n = 30) serum samples with a wide range of anti-SARS-CoV-2 antibody titers and negative ( n = 80) plasma and/or serum samples that were collected before the start of the COVID-19 pandemic. Each sample was characterized for anti-SARS-CoV-2 antibodies against the spike protein using enzyme-linked immunosorbent assays (ELISA). Samples were selected for the panel when there was agreement on seropositivity by laboratories at National Cancer Institute's Frederick National Laboratory for Cancer Research (NCI-FNLCR) and Centers for Disease Control and Prevention (CDC). The sensitivity and specificity of each assay were assessed to determine Emergency Use Authorization (EUA) suitability. As of January 8, 2021, results from 91 evaluations were made publicly available (https://open.fda.gov/apis/device/covid19serology/, and https://www.cdc.gov/coronavirus/2019-ncov/covid-data/serology-surveillance/serology-test-evaluation.html). Sensitivity ranged from 27% to 100% for IgG ( n = 81), from 10% to 100% for IgM ( n = 74), and from 73% to 100% for total or pan-immunoglobulins ( n = 5). The combined specificity ranged from 58% to 100% ( n = 91). Approximately one-third ( n = 27) of the assays evaluated are now authorized by FDA for emergency use. This collaboration established a framework for assay performance evaluation that could be used for future outbreaks and could serve as a model for other technologies. IMPORTANCE The SARS-CoV-2 pandemic created a crucial need for accurate serology assays to evaluate seroprevalence and antiviral immune responses. The initial flood of serology assays entering the market with inadequate performance emphasized the need for independent evaluation of commercial SARS-CoV-2 antibody assays using performance evaluation panels to determine suitability for use under EUA. Through a government-wide collaborative network, 91 commercial SARS-CoV-2 serology assay evaluations were performed. Three evaluation panels with similar overall antibody titer distributions were assembled to evaluate performance. Nearly one-third of the assays evaluated met acceptable performance recommendations, and two assays had EUAs revoked and were removed from the U.S. market based on inadequate performance. Data for all serology assays evaluated are available at the FDA and CDC websites (https://open.fda.gov/apis/device/covid19serology/, and https://www.cdc.gov/coronavirus/2019-ncov/covid-data/serology-surveillance/serology-test-evaluation.html).
- Published
- 2022
- Full Text
- View/download PDF
21. Therapy-Induced Senescence: Opportunities to Improve Anticancer Therapy.
- Author
-
Prasanna PG, Citrin DE, Hildesheim J, Ahmed MM, Venkatachalam S, Riscuta G, Xi D, Zheng G, Deursen JV, Goronzy J, Kron SJ, Anscher MS, Sharpless NE, Campisi J, Brown SL, Niedernhofer LJ, O'Loghlen A, Georgakilas AG, Paris F, Gius D, Gewirtz DA, Schmitt CA, Abazeed ME, Kirkland JL, Richmond A, Romesser PB, Lowe SW, Gil J, Mendonca MS, Burma S, Zhou D, and Coleman CN
- Subjects
- Biomarkers, Humans, Senescence-Associated Secretory Phenotype, Cellular Senescence, Neoplasms drug therapy, Neoplasms pathology
- Abstract
Cellular senescence is an essential tumor suppressive mechanism that prevents the propagation of oncogenically activated, genetically unstable, and/or damaged cells. Induction of tumor cell senescence is also one of the underlying mechanisms by which cancer therapies exert antitumor activity. However, an increasing body of evidence from preclinical studies demonstrates that radiation and chemotherapy cause accumulation of senescent cells (SnCs) both in tumor and normal tissue. SnCs in tumors can, paradoxically, promote tumor relapse, metastasis, and resistance to therapy, in part, through expression of the senescence-associated secretory phenotype. In addition, SnCs in normal tissue can contribute to certain radiation- and chemotherapy-induced side effects. Because of its multiple roles, cellular senescence could serve as an important target in the fight against cancer. This commentary provides a summary of the discussion at the National Cancer Institute Workshop on Radiation, Senescence, and Cancer (August 10-11, 2020, National Cancer Institute, Bethesda, MD) regarding the current status of senescence research, heterogeneity of therapy-induced senescence, current status of senotherapeutics and molecular biomarkers, a concept of "one-two punch" cancer therapy (consisting of therapeutics to induce tumor cell senescence followed by selective clearance of SnCs), and its integration with personalized adaptive tumor therapy. It also identifies key knowledge gaps and outlines future directions in this emerging field to improve treatment outcomes for cancer patients., (© The Author(s) 2021. Published by Oxford University Press.)
- Published
- 2021
- Full Text
- View/download PDF
22. Cancer as a Global Health Priority.
- Author
-
Gopal S and Sharpless NE
- Published
- 2021
- Full Text
- View/download PDF
23. The potential of AI in cancer care and research.
- Author
-
Sharpless NE and Kerlavage AR
- Subjects
- Animals, Diffusion of Innovation, Humans, Machine Learning, United States, Artificial Intelligence, Biomedical Research, Medical Oncology, National Cancer Institute (U.S.), Neoplasms diagnostic imaging, Neoplasms genetics, Neoplasms metabolism, Neoplasms therapy
- Abstract
Current applications of artificial intelligence (AI), machine learning, and deep learning in cancer research and clinical care are highly diverse-from aiding radiologists in reading medical images to predicting oncoprotein folding and dynamics. The list of available AI-based tools is growing rapidly and will only continue to expand. With the immense potential for AI to advance cancer research and clinical care, the National Cancer Institute (NCI) has a responsibility to consider and support the development and evaluation of such technologies. NCI's current involvement in AI research spans the spectrum of development, implementation, and assessment. That includes generating large, publicly available, curated datasets; shifting the culture of data sharing; training the next generation of scientists in both AI and cancer sciences; fostering interdisciplinary collaborations; investing in research to improve AI methods and models that are designed specifically for cancer; widening access to computing power; procuring computer architecture for future developments; and assuring AI research and technologies follow ethical principles. In addition to a broad overview of AI applications in cancer research and care, and NCI's ongoing AI-based activities, this Perspective outlines NCI's four priority areas for future investment of cancer-focused AI development., (Published by Elsevier B.V.)
- Published
- 2021
- Full Text
- View/download PDF
24. Cancer Informatics for Cancer Centers: Scientific Drivers for Informatics, Data Science, and Care in Pediatric, Adolescent, and Young Adult Cancer.
- Author
-
Kerlavage AR, Kirchhoff AC, Guidry Auvil JM, Sharpless NE, Davis KL, Reilly K, Reaman G, Penberthy L, Deapen D, Hwang A, Durbin EB, Gallotto SL, Aplenc R, Volchenboum SL, Heath AP, Aronow BJ, Zhang J, Vaske O, Alonzo TA, Nathan PC, Poynter JN, Armstrong G, Hahn EE, Wernli KJ, Greene C, DiGiovanna J, Resnick AC, Shalley ER, Nadaf S, and Kibbe WA
- Subjects
- Adolescent, Child, Data Science, Humans, Pandemics, SARS-CoV-2, Young Adult, COVID-19, Medical Informatics, Neoplasms epidemiology, Neoplasms therapy
- Abstract
Cancer Informatics for Cancer Centers (CI4CC) is a grassroots, nonprofit 501c3 organization intended to provide a focused national forum for engagement of senior cancer informatics leaders, primarily aimed at academic cancer centers anywhere in the world but with a special emphasis on the 70 National Cancer Institute-funded cancer centers. This consortium has regularly held topic-focused biannual face-to-face symposiums. These meetings are a place to review cancer informatics and data science priorities and initiatives, providing a forum for discussion of the strategic and pragmatic issues that we faced at our respective institutions and cancer centers. Here, we provide meeting highlights from the latest CI4CC Symposium, which was delayed from its original April 2020 schedule because of the COVID-19 pandemic and held virtually over three days (September 24, October 1, and October 8) in the fall of 2020. In addition to the content presented, we found that holding this event virtually once a week for 6 hours was a great way to keep the kind of deep engagement that a face-to-face meeting engenders. This is the second such publication of CI4CC Symposium highlights, the first covering the meeting that took place in Napa, California, from October 14-16, 2019. We conclude with some thoughts about using data science to learn from every child with cancer, focusing on emerging activities of the National Cancer Institute's Childhood Cancer Data Initiative., Competing Interests: Anne C. KirchhoffStock and Other Ownership Interests: Medtronic Kara L. DavisHonoraria: NovartisResearch Funding: Jazz Pharmaceuticals Karlyne ReillyConsulting or Advisory Role: Saul Ewing LLCPatents, Royalties, Other Intellectual Property: I hold a patent to a potential therapeutic: Reilly KM, Beutler JA, Turbyville T, Wiemer DF: The Natural Product Schweinfurthin A and Synthetic Schweinfurthin Analogs Specifically Inhibit Nf1-Null Cells and may be Useful as a Therapy for Neurofibromatosis Type 1. US patent 61/174,338, April 19, 2014; International patent PCT/US10/33153. There are no royalties or licensing fees from this patent at the time Richard AplencExpert Testimony: Vorys Samuel L. VolchenboumStock and Other Ownership Interests: Litmus HealthConsulting or Advisory Role: AccordantTravel, Accommodations, Expenses: Sanford Health Bruce J. AronowPatents, Royalties, Other Intellectual Property: Patents issued for some data analysis algorithms related to data mining and discovery approaches to drug repositioning for new clinical indications Olena VaskeEmployment: NantWorksStock and Other Ownership Interests: NantHealth Casey GreeneOther Relationship: Alex's Lemonade Stand Foundation Jack DiGiovannaEmployment: Biogen (I)Stock and Other Ownership Interests: Biogen Sorena NadafThis author is a member of the JCO Clinical Cancer Informatics Editorial Board. Journal policy recused the author from having any role in the peer review of this manuscript. Warren A. KibbeThis author is a member of the JCO Clinical Cancer Informatics Editorial Board. Journal policy recused the author from having any role in the peer review of this manuscript.No other potential conflicts of interest were reported.
- Published
- 2021
- Full Text
- View/download PDF
25. Progress and potential: The Cancer Moonshot.
- Author
-
Sharpless NE and Singer DS
- Subjects
- Humans, Precision Medicine, United States, Biomedical Research economics, Biomedical Research legislation & jurisprudence, Government Programs, Health Policy, Healthcare Disparities, Neoplasms diagnosis, Neoplasms therapy
- Published
- 2021
- Full Text
- View/download PDF
26. Association of SARS-CoV-2 Seropositive Antibody Test With Risk of Future Infection.
- Author
-
Harvey RA, Rassen JA, Kabelac CA, Turenne W, Leonard S, Klesh R, Meyer WA 3rd, Kaufman HW, Anderson S, Cohen O, Petkov VI, Cronin KA, Van Dyke AL, Lowy DR, Sharpless NE, and Penberthy LT
- Subjects
- Adult, Age Factors, Antibodies, Viral isolation & purification, Correlation of Data, Female, Humans, Male, Middle Aged, Seroepidemiologic Studies, Symptom Assessment methods, Symptom Assessment statistics & numerical data, United States epidemiology, Virus Shedding immunology, COVID-19 blood, COVID-19 diagnosis, COVID-19 epidemiology, COVID-19 prevention & control, COVID-19 Nucleic Acid Testing methods, COVID-19 Nucleic Acid Testing statistics & numerical data, COVID-19 Serological Testing methods, COVID-19 Serological Testing statistics & numerical data, Disease Susceptibility diagnosis, Disease Susceptibility epidemiology, Disease Susceptibility immunology, SARS-CoV-2 immunology, SARS-CoV-2 isolation & purification
- Abstract
Importance: Understanding the effect of serum antibodies to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) on susceptibility to infection is important for identifying at-risk populations and could have implications for vaccine deployment., Objective: The study purpose was to evaluate evidence of SARS-CoV-2 infection based on diagnostic nucleic acid amplification test (NAAT) among patients with positive vs negative test results for antibodies in an observational descriptive cohort study of clinical laboratory and linked claims data., Design, Setting, and Participants: The study created cohorts from a deidentified data set composed of commercial laboratory tests, medical and pharmacy claims, electronic health records, and hospital chargemaster data. Patients were categorized as antibody-positive or antibody-negative according to their first SARS-CoV-2 antibody test in the database., Main Outcomes and Measures: Primary end points were post-index diagnostic NAAT results, with infection defined as a positive diagnostic test post-index, measured in 30-day intervals (0-30, 31-60, 61-90, >90 days). Additional measures included demographic, geographic, and clinical characteristics at the time of the index antibody test, including recorded signs and symptoms or prior evidence of coronavirus 2019 (COVID) diagnoses or positive NAAT results and recorded comorbidities., Results: The cohort included 3 257 478 unique patients with an index antibody test; 56% were female with a median (SD) age of 48 (20) years. Of these, 2 876 773 (88.3%) had a negative index antibody result, and 378 606 (11.6%) had a positive index antibody result. Patients with a negative antibody test result were older than those with a positive result (mean age 48 vs 44 years). Of index-positive patients, 18.4% converted to seronegative over the follow-up period. During the follow-up periods, the ratio (95% CI) of positive NAAT results among individuals who had a positive antibody test at index vs those with a negative antibody test at index was 2.85 (95% CI, 2.73-2.97) at 0 to 30 days, 0.67 (95% CI, 0.6-0.74) at 31 to 60 days, 0.29 (95% CI, 0.24-0.35) at 61 to 90 days, and 0.10 (95% CI, 0.05-0.19) at more than 90 days., Conclusions and Relevance: In this cohort study, patients with positive antibody test results were initially more likely to have positive NAAT results, consistent with prolonged RNA shedding, but became markedly less likely to have positive NAAT results over time, suggesting that seropositivity is associated with protection from infection. The duration of protection is unknown, and protection may wane over time.
- Published
- 2021
- Full Text
- View/download PDF
27. Corrigendum to "Role of senescence marker p16 INK4A measured in peripheral blood T-lymphocytes in predicting length of hospital stay after coronary artery bypass surgery in older adults" [Exp. Gerontol. 74 (2016) 29-36].
- Author
-
Pustavoitau A, Barodka V, Sharpless NE, Torrice C, Nyhan D, Berkowitz DE, Shah AS, Bandeen Roche KJ, and Walston J
- Published
- 2021
- Full Text
- View/download PDF
28. Cancer Grand Challenges: Embarking on a New Era of Discovery.
- Author
-
Foulkes I and Sharpless NE
- Subjects
- Humans, Biomedical Research methods, Neoplasms epidemiology
- Abstract
Cancer Grand Challenges is a unique funding platform that dares global, multidisciplinary teams of researchers to come together, think differently, and tackle some of the toughest challenges in cancer research. Here, we discuss the nine intractable challenges currently open for application., (©2020 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
29. Real-world data suggest antibody positivity to SARS-CoV-2 is associated with a decreased risk of future infection.
- Author
-
Harvey RA, Rassen JA, Kabelac CA, Turenne W, Leonard S, Klesh R, Meyer WA 3rd, Kaufman HW, Anderson S, Cohen O, Petkov VI, Cronin KA, Van Dyke AL, Lowy DR, Sharpless NE, and Penberthy LT
- Abstract
Importance There is limited evidence regarding whether the presence of serum antibodies to SARS-CoV-2 is associated with a decreased risk of future infection. Understanding susceptibility to infection and the role of immune memory is important for identifying at-risk populations and could have implications for vaccine deployment. Objective The purpose of this study was to evaluate subsequent evidence of SARS-CoV-2 infection based on diagnostic nucleic acid amplification test (NAAT) among individuals who are antibody-positive compared with those who are antibody-negative, using real-world data. Design This was an observational descriptive cohort study. Participants The study utilized a national sample to create cohorts from a de-identified dataset composed of commercial laboratory test results, open and closed medical and pharmacy claims, electronic health records, hospital billing (chargemaster) data, and payer enrollment files from the United States. Patients were indexed as antibody-positive or antibody-negative according to their first SARS-CoV-2 antibody test recorded in the database. Patients with more than 1 antibody test on the index date where results were discordant were excluded. Main Outcomes/Measures Primary endpoints were index antibody test results and post-index diagnostic NAAT results, with infection defined as a positive diagnostic test post-index, as measured in 30-day intervals (0-30, 31-60, 61-90, >90 days). Additional measures included demographic, geographic, and clinical characteristics at the time of the index antibody test, such as recorded signs and symptoms or prior evidence of COVID-19 (diagnoses or NAAT+) and recorded comorbidities. Results We included 3,257,478 unique patients with an index antibody test. Of these, 2,876,773 (88.3%) had a negative index antibody result, 378,606 (11.6%) had a positive index antibody result, and 2,099 (0.1%) had an inconclusive index antibody result. Patients with a negative antibody test were somewhat older at index than those with a positive result (mean of 48 versus 44 years). A fraction (18.4%) of individuals who were initially seropositive converted to seronegative over the follow up period. During the follow-up periods, the ratio (CI) of positive NAAT results among individuals who had a positive antibody test at index versus those with a negative antibody test at index was 2.85 (2.73 - 2.97) at 0-30 days, 0.67 (0.6 - 0.74) at 31-60 days, 0.29 (0.24 - 0.35) at 61-90 days), and 0.10 (0.05 - 0.19) at >90 days. Conclusions Patients who display positive antibody tests are initially more likely to have a positive NAAT, consistent with prolonged RNA shedding, but over time become markedly less likely to have a positive NAAT. This result suggests seropositivity using commercially available assays is associated with protection from infection. The duration of protection is unknown and may wane over time; this parameter will need to be addressed in a study with extended duration of follow up.
- Published
- 2020
- Full Text
- View/download PDF
30. Redoubling Efforts to Help Americans Quit Smoking - Federal Initiatives to Tackle the Country's Longest-Running Epidemic.
- Author
-
Redfield RR, Hahn SM, and Sharpless NE
- Subjects
- Humans, Physician's Role, Smoking adverse effects, Smoking mortality, State Government, United States epidemiology, United States Public Health Service, Health Promotion methods, Smoking epidemiology, Smoking Cessation methods
- Published
- 2020
- Full Text
- View/download PDF
31. Advancing progress for patients with cancer through small business innovation research.
- Author
-
Sharpless NE
- Subjects
- Humans, Biomedical Research, Neoplasms, Small Business
- Published
- 2020
- Full Text
- View/download PDF
32. COVID-19 and cancer.
- Author
-
Sharpless NE
- Subjects
- Betacoronavirus, COVID-19, Coronavirus Infections epidemiology, Forecasting, Humans, Models, Theoretical, Neoplasms mortality, Pandemics, Pneumonia, Viral epidemiology, SARS-CoV-2, United States epidemiology, Breast Neoplasms mortality, Colorectal Neoplasms mortality, Coronavirus Infections complications, Pneumonia, Viral complications
- Published
- 2020
- Full Text
- View/download PDF
33. Cells exhibiting strong p16 INK4a promoter activation in vivo display features of senescence.
- Author
-
Liu JY, Souroullas GP, Diekman BO, Krishnamurthy J, Hall BM, Sorrentino JA, Parker JS, Sessions GA, Gudkov AV, and Sharpless NE
- Subjects
- Animals, Cell Proliferation, Enzyme Activation, Fibroblasts metabolism, Half-Life, Humans, Mice, Phenotype, RNA, Messenger genetics, beta-Galactosidase metabolism, Cellular Senescence genetics, Cyclin-Dependent Kinase Inhibitor p16 genetics, Promoter Regions, Genetic
- Abstract
The activation of cellular senescence throughout the lifespan promotes tumor suppression, whereas the persistence of senescent cells contributes to aspects of aging. This theory has been limited, however, by an inability to identify and isolate individual senescent cells within an intact organism. Toward that end, we generated a murine reporter strain by "knocking-in" a fluorochrome, tandem-dimer Tomato (tdTom), into exon 1α of the p16
INK4a locus. We used this allele ( p16tdTom ) for the enumeration, isolation, and characterization of individual p16INK4a -expressing cells (tdTom+ ). The half-life of the knocked-in transcript was shorter than that of the endogenous p16INK4a mRNA, and therefore reporter expression better correlated with p16INK4a promoter activation than p16INK4a transcript abundance. The frequency of tdTom+ cells increased with serial passage in cultured murine embryo fibroblasts from p16tdTom/+ mice. In adult mice, tdTom+ cells could be readily detected at low frequency in many tissues, and the frequency of these cells increased with aging. Using an in vivo model of peritoneal inflammation, we compared the phenotype of cells with or without activation of p16INK4a and found that tdTom+ macrophages exhibited some features of senescence, including reduced proliferation, senescence-associated β-galactosidase (SA-β-gal) activation, and increased mRNA expression of a subset of transcripts encoding factors involved in SA-secretory phenotype (SASP). These results indicate that cells harboring activation of the p16INK4a promoter accumulate with aging and inflammation in vivo, and display characteristics of senescence., Competing Interests: The authors declare no conflict of interest., (Copyright © 2019 the Author(s). Published by PNAS.)- Published
- 2019
- Full Text
- View/download PDF
34. Modernizing Clinical Trials for Patients With Cancer.
- Author
-
Sharpless NE and Doroshow JH
- Subjects
- Drug Industry, Humans, Intersectoral Collaboration, National Cancer Institute (U.S.), Patient Selection, Research Support as Topic, United States, Clinical Trials as Topic economics, Clinical Trials as Topic organization & administration, Neoplasms therapy
- Published
- 2019
- Full Text
- View/download PDF
35. The Prognostic Significance of Low-Frequency Somatic Mutations in Metastatic Cutaneous Melanoma.
- Author
-
Zhao X, Little P, Hoyle AP, Pegna GJ, Hayward MC, Ivanova A, Parker JS, Marron DL, Soloway MG, Jo H, Salazar AH, Papakonstantinou MP, Bouchard DM, Jefferys SR, Hoadley KA, Ollila DW, Frank JS, Thomas NE, Googe PB, Ezzell AJ, Collichio FA, Lee CB, Earp HS, Sharpless NE, Hugo W, Wilmott JS, Quek C, Waddell N, Johansson PA, Thompson JF, Hayward NK, Mann GJ, Lo RS, Johnson DB, Scolyer RA, Hayes DN, and Moschos SJ
- Abstract
Background: Little is known about the prognostic significance of somatically mutated genes in metastatic melanoma (MM). We have employed a combined clinical and bioinformatics approach on tumor samples from cutaneous melanoma (SKCM) as part of The Cancer Genome Atlas project (TCGA) to identify mutated genes with potential clinical relevance. Methods: After limiting our DNA sequencing analysis to MM samples ( n = 356) and to the CANCER CENSUS gene list, we filtered out mutations with low functional significance (snpEFF). We performed Cox analysis on 53 genes that were mutated in ≥3% of samples, and had ≥50% difference in incidence of mutations in deceased subjects versus alive subjects. Results: Four genes were potentially prognostic [ RAC1, FGFR1, CARD11, CIITA ; false discovery rate (FDR) < 0.2]. We identified 18 additional genes (e.g., SPEN, PDGFRB, GNAS, MAP2K1, EGFR, TSC2 ) that were less likely to have prognostic value (FDR < 0.4). Most somatic mutations in these 22 genes were infrequent (< 10%), associated with high somatic mutation burden, and were evenly distributed across all exons, except for RAC1 and MAP2K1 . Mutations in only 9 of these 22 genes were also identified by RNA sequencing in >75% of the samples that exhibited corresponding DNA mutations. The low frequency, UV signature type and RNA expression of the 22 genes in MM samples were confirmed in a separate multi-institution validation cohort ( n = 413). An underpowered analysis within a subset of this validation cohort with available patient follow-up ( n = 224) showed that somatic mutations in SPEN and RAC1 reached borderline prognostic significance [log-rank favorable ( p = 0.09) and adverse ( p = 0.07), respectively]. Somatic mutations in SPEN , and to a lesser extent RAC1 , were not associated with definite gene copy number or RNA expression alterations. High (>2+) nuclear plus cytoplasmic expression intensity for SPEN was associated with longer melanoma-specific overall survival (OS) compared to lower (≤ 2+) nuclear intensity ( p = 0.048). We conclude that expressed somatic mutations in infrequently mutated genes beyond the well-characterized ones (e.g., BRAF, RAS, CDKN2A, PTEN, TP53 ), such as RAC1 and SPEN , may have prognostic significance in MM.
- Published
- 2019
- Full Text
- View/download PDF
36. Expression of p16 INK 4a is a biomarker of chondrocyte aging but does not cause osteoarthritis.
- Author
-
Diekman BO, Sessions GA, Collins JA, Knecht AK, Strum SL, Mitin NK, Carlson CS, Loeser RF, and Sharpless NE
- Subjects
- Aged, Animals, Biomarkers analysis, Biomarkers metabolism, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Cell Proliferation drug effects, Cellular Senescence drug effects, Chondrocytes drug effects, Cyclic N-Oxides, Cyclin-Dependent Kinase Inhibitor p16 antagonists & inhibitors, Cyclin-Dependent Kinase Inhibitor p16 metabolism, Cyclin-Dependent Kinases antagonists & inhibitors, Cyclin-Dependent Kinases metabolism, Humans, Indolizines, Male, Mice, Mice, Inbred C57BL, Middle Aged, Osteoarthritis genetics, Osteoarthritis metabolism, Piperazines pharmacology, Protein Kinase Inhibitors pharmacology, Pyridines pharmacology, Pyridinium Compounds pharmacology, RNA, Small Interfering pharmacology, Young Adult, Cellular Senescence genetics, Chondrocytes metabolism, Cyclin-Dependent Kinase Inhibitor p16 genetics
- Abstract
Cellular senescence drives a functional decline of numerous tissues with aging by limiting regenerative proliferation and/or by producing pro-inflammatory molecules known as the senescence-associated secretory phenotype (SASP). The senescence biomarker p16
INK 4a is a potent inhibitor of the cell cycle but is not essential for SASP production. Thus, it is unclear whether p16INK 4a identifies senescence in hyporeplicative cells such as articular chondrocytes and whether p16INK 4a contributes to pathologic characteristics of cartilage aging. To address these questions, we examined the role of p16INK 4a in murine and human models of chondrocyte aging. We observed that p16INK 4a mRNA expression was significantly upregulated with chronological aging in murine cartilage (~50-fold from 4 to 18 months of age) and in primary human chondrocytes from 57 cadaveric donors (r2 = .27, p < .0001). Human chondrocytes exhibited substantial replicative potential in vitro that depended on the activity of cyclin-dependent kinases 4 or 6 (CDK4/6), and proliferation was reduced in cells from older donors with increased p16INK 4a expression. Moreover, increased chondrocyte p16INK 4a expression correlated with several SASP transcripts. Despite the relationship between p16INK 4a expression and these features of senescence, somatic inactivation of p16INK 4a in chondrocytes of adult mice did not mitigate SASP expression and did not alter the rate of osteoarthritis (OA) with physiological aging or after destabilization of the medial meniscus. These results establish that p16INK 4a expression is a biomarker of dysfunctional chondrocytes, but that the effects of chondrocyte senescence on OA are more likely driven by production of SASP molecules than by loss of chondrocyte replicative function., (© 2018 The Authors. Aging Cell published by the Anatomical Society and John Wiley & Sons Ltd.)- Published
- 2018
- Full Text
- View/download PDF
37. CDK4/6 Inhibition Augments Antitumor Immunity by Enhancing T-cell Activation.
- Author
-
Deng J, Wang ES, Jenkins RW, Li S, Dries R, Yates K, Chhabra S, Huang W, Liu H, Aref AR, Ivanova E, Paweletz CP, Bowden M, Zhou CW, Herter-Sprie GS, Sorrentino JA, Bisi JE, Lizotte PH, Merlino AA, Quinn MM, Bufe LE, Yang A, Zhang Y, Zhang H, Gao P, Chen T, Cavanaugh ME, Rode AJ, Haines E, Roberts PJ, Strum JC, Richards WG, Lorch JH, Parangi S, Gunda V, Boland GM, Bueno R, Palakurthi S, Freeman GJ, Ritz J, Haining WN, Sharpless NE, Arthanari H, Shapiro GI, Barbie DA, Gray NS, and Wong KK
- Subjects
- Animals, Antineoplastic Agents pharmacology, Antineoplastic Agents, Immunological pharmacology, Cell Line, Tumor, Humans, Lymphocytes, Tumor-Infiltrating drug effects, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating metabolism, Mice, Neoplasms diagnosis, Neoplasms genetics, Neoplasms metabolism, Protein Kinase Inhibitors pharmacology, T-Lymphocytes metabolism, Xenograft Model Antitumor Assays, Cyclin-Dependent Kinase 4 antagonists & inhibitors, Cyclin-Dependent Kinase 6 antagonists & inhibitors, Lymphocyte Activation drug effects, Lymphocyte Activation immunology, Neoplasms immunology, T-Lymphocytes drug effects, T-Lymphocytes immunology
- Abstract
Immune checkpoint blockade, exemplified by antibodies targeting the PD-1 receptor, can induce durable tumor regressions in some patients. To enhance the efficacy of existing immunotherapies, we screened for small molecules capable of increasing the activity of T cells suppressed by PD-1. Here, we show that short-term exposure to small-molecule inhibitors of cyclin-dependent kinases 4 and 6 (CDK4/6) significantly enhances T-cell activation, contributing to antitumor effects in vivo , due in part to the derepression of NFAT family proteins and their target genes, critical regulators of T-cell function. Although CDK4/6 inhibitors decrease T-cell proliferation, they increase tumor infiltration and activation of effector T cells. Moreover, CDK4/6 inhibition augments the response to PD-1 blockade in a novel ex vivo organotypic tumor spheroid culture system and in multiple in vivo murine syngeneic models, thereby providing a rationale for combining CDK4/6 inhibitors and immunotherapies. Significance: Our results define previously unrecognized immunomodulatory functions of CDK4/6 and suggest that combining CDK4/6 inhibitors with immune checkpoint blockade may increase treatment efficacy in patients. Furthermore, our study highlights the critical importance of identifying complementary strategies to improve the efficacy of immunotherapy for patients with cancer. Cancer Discov; 8(2); 216-33. ©2017 AACR. See related commentary by Balko and Sosman, p. 143 See related article by Jenkins et al., p. 196 This article is highlighted in the In This Issue feature, p. 127 ., (©2017 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
38. Enhancing Next-Generation Sequencing-Guided Cancer Care Through Cognitive Computing.
- Author
-
Patel NM, Michelini VV, Snell JM, Balu S, Hoyle AP, Parker JS, Hayward MC, Eberhard DA, Salazar AH, McNeillie P, Xu J, Huettner CS, Koyama T, Utro F, Rhrissorrakrai K, Norel R, Bilal E, Royyuru A, Parida L, Earp HS, Grilley-Olson JE, Hayes DN, Harvey SJ, Sharpless NE, and Kim WY
- Subjects
- Biomarkers, Tumor, Case-Control Studies, Combined Modality Therapy, Follow-Up Studies, Gene Expression Regulation, Neoplastic, High-Throughput Nucleotide Sequencing, Humans, Lymphatic Metastasis, Neoplasm Invasiveness, Neoplasm Recurrence, Local drug therapy, Neoplasm Recurrence, Local pathology, Neoplasms pathology, Prognosis, Retrospective Studies, Survival Rate, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Neoplasms drug therapy
- Abstract
Background: Using next-generation sequencing (NGS) to guide cancer therapy has created challenges in analyzing and reporting large volumes of genomic data to patients and caregivers. Specifically, providing current, accurate information on newly approved therapies and open clinical trials requires considerable manual curation performed mainly by human "molecular tumor boards" (MTBs). The purpose of this study was to determine the utility of cognitive computing as performed by Watson for Genomics (WfG) compared with a human MTB., Materials and Methods: One thousand eighteen patient cases that previously underwent targeted exon sequencing at the University of North Carolina (UNC) and subsequent analysis by the UNCseq informatics pipeline and the UNC MTB between November 7, 2011, and May 12, 2015, were analyzed with WfG, a cognitive computing technology for genomic analysis., Results: Using a WfG-curated actionable gene list, we identified additional genomic events of potential significance (not discovered by traditional MTB curation) in 323 (32%) patients. The majority of these additional genomic events were considered actionable based upon their ability to qualify patients for biomarker-selected clinical trials. Indeed, the opening of a relevant clinical trial within 1 month prior to WfG analysis provided the rationale for identification of a new actionable event in nearly a quarter of the 323 patients. This automated analysis took <3 minutes per case., Conclusion: These results demonstrate that the interpretation and actionability of somatic NGS results are evolving too rapidly to rely solely on human curation. Molecular tumor boards empowered by cognitive computing could potentially improve patient care by providing a rapid, comprehensive approach for data analysis and consideration of up-to-date availability of clinical trials., Implications for Practice: The results of this study demonstrate that the interpretation and actionability of somatic next-generation sequencing results are evolving too rapidly to rely solely on human curation. Molecular tumor boards empowered by cognitive computing can significantly improve patient care by providing a fast, cost-effective, and comprehensive approach for data analysis in the delivery of precision medicine. Patients and physicians who are considering enrollment in clinical trials may benefit from the support of such tools applied to genomic data., Competing Interests: Disclosures of potential conflicts of interest may be found at the end of this article., (© AlphaMed Press 2017.)
- Published
- 2018
- Full Text
- View/download PDF
39. New Mechanisms of Resistance to MEK Inhibitors in Melanoma Revealed by Intravital Imaging.
- Author
-
Brighton HE, Angus SP, Bo T, Roques J, Tagliatela AC, Darr DB, Karagoz K, Sciaky N, Gatza ML, Sharpless NE, Johnson GL, and Bear JE
- Subjects
- Animals, Apoptosis drug effects, Case-Control Studies, Cell Proliferation drug effects, Gene Expression Profiling, Humans, Longitudinal Studies, Melanoma drug therapy, Melanoma metabolism, Mice, Mutation, Prognosis, Signal Transduction, Tumor Cells, Cultured, Biomarkers, Tumor genetics, Drug Resistance, Neoplasm, Intravital Microscopy methods, MAP Kinase Kinase 1 antagonists & inhibitors, Melanoma pathology, Protein Kinase Inhibitors pharmacology, Pyridones pharmacology, Pyrimidinones pharmacology
- Abstract
Targeted therapeutics that are initially effective in cancer patients nearly invariably engender resistance at some stage, an inherent challenge in the use of any molecular-targeted drug in cancer settings. In this study, we evaluated resistance mechanisms arising in metastatic melanoma to MAPK pathway kinase inhibitors as a strategy to identify candidate strategies to limit risks of resistance. To investigate longitudinal responses, we developed an intravital serial imaging approach that can directly visualize drug response in an inducible RAF-driven, autochthonous murine model of melanoma incorporating a fluorescent reporter allele (tdTomatoLSL). Using this system, we visualized formation and progression of tumors in situ , starting from the single-cell level longitudinally over time. Reliable reporting of the status of primary murine tumors treated with the selective MEK1/2 inhibitor (MEKi) trametinib illustrated a time-course of initial drug response and persistence, followed by the development of drug resistance. We found that tumor cells adjacent to bundled collagen had a preferential persistence in response to MEKi. Unbiased transcriptional and kinome reprogramming analyses from selected treatment time points suggested increased c-Kit and PI3K/AKT pathway activation in resistant tumors, along with enhanced expression of epithelial genes and epithelial-mesenchymal transition downregulation signatures with development of MEKi resistance. Similar trends were observed following simultaneous treatment with BRAF and MEK inhibitors aligned to standard-of-care combination therapy, suggesting these reprogramming events were not specific to MEKi alone. Overall, our results illuminate the integration of tumor-stroma dynamics with tissue plasticity in melanoma progression and provide new insights into the basis for drug response, persistence, and resistance. Significance: A longitudinal study tracks the course of MEKi treatment in an autochthonous imageable murine model of melanoma from initial response to therapeutic resistance, offering new insights into the basis for drug response, persistence, and resistance. Cancer Res; 78(2); 542-57. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
40. Exploiting Drug Addiction Mechanisms to Select against MAPKi-Resistant Melanoma.
- Author
-
Hong A, Moriceau G, Sun L, Lomeli S, Piva M, Damoiseaux R, Holmen SL, Sharpless NE, Hugo W, and Lo RS
- Subjects
- Animals, Humans, Melanoma pathology, Mice, Protein Kinase Inhibitors pharmacology, Drug Resistance, Neoplasm genetics, Melanoma drug therapy, Protein Kinase Inhibitors therapeutic use, Substance-Related Disorders genetics
- Abstract
Melanoma resistant to MAPK inhibitors (MAPKi) displays loss of fitness upon experimental MAPKi withdrawal and, clinically, may be resensitized to MAPKi therapy after a drug holiday. Here, we uncovered and therapeutically exploited the mechanisms of MAPKi addiction in MAPKi-resistant BRAF
MUT or NRASMUT melanoma. MAPKi-addiction phenotypes evident upon drug withdrawal spanned transient cell-cycle slowdown to cell-death responses, the latter of which required a robust phosphorylated ERK (pERK) rebound. Generally, drug withdrawal-induced pERK rebound upregulated p38-FRA1-JUNB-CDKN1A and downregulated proliferation, but only a robust pERK rebound resulted in DNA damage and parthanatos-related cell death. Importantly, pharmacologically impairing DNA damage repair during MAPKi withdrawal augmented MAPKi addiction across the board by converting a cell-cycle deceleration to a caspase-dependent cell-death response or by furthering parthanatos-related cell death. Specifically in MEKi-resistant NRASMUT or atypical BRAFMUT melanoma, treatment with a type I RAF inhibitor intensified pERK rebound elicited by MEKi withdrawal, thereby promoting a cell death-predominant MAPKi-addiction phenotype. Thus, MAPKi discontinuation upon disease progression should be coupled with specific strategies that augment MAPKi addiction. Significance: Discontinuing targeted therapy may select against drug-resistant tumor clones, but drug-addiction mechanisms are ill-defined. Using melanoma resistant to but withdrawn from MAPKi, we defined a synthetic lethality between supraphysiologic levels of pERK and DNA damage. Actively promoting this synthetic lethality could rationalize sequential/rotational regimens that address evolving vulnerabilities. Cancer Discov; 8(1); 74-93. ©2017 AACR. See related commentary by Stern, p. 20 This article is highlighted in the In This Issue feature, p. 1 ., (©2017 American Association for Cancer Research.)- Published
- 2018
- Full Text
- View/download PDF
41. Constitutive Ras signaling and Ink4a/Arf inactivation cooperate during the development of B-ALL in mice.
- Author
-
Sewastianik T, Jiang M, Sukhdeo K, Patel SS, Roberts K, Kang Y, Alduaij A, Dennis PS, Lawney B, Liu R, Song Z, Xiong J, Zhang Y, Lemieux ME, Pinkus GS, Rich JN, Weinstock DM, Mullighan CG, Sharpless NE, and Carrasco RD
- Abstract
Despite recent advances in treatment, human precursor B-cell acute lymphoblastic leukemia (B-ALL) remains a challenging clinical entity. Recent genome-wide studies have uncovered frequent genetic alterations involving RAS pathway mutations and loss of the INK4A /ARF locus, suggesting their important role in the pathogenesis, relapse, and chemotherapy resistance of B-ALL. To better understand the oncogenic mechanisms by which these alterations might promote B-ALL and to develop an in vivo preclinical model of relapsed B-ALL, we engineered mouse strains with induced somatic Kras
G12D pathway activation and/or loss of Ink4a/Arf during early stages of B-cell development. Although constitutive activation of KrasG12D in B cells induced prominent transcriptional changes that resulted in enhanced proliferation, it was not sufficient by itself to induce development of a high-grade leukemia/lymphoma. Instead, in 40% of mice, these engineered mutations promoted development of a clonal low-grade lymphoproliferative disorder resembling human extranodal marginal-zone lymphoma of mucosa-associated lymphoid tissue or lymphoplasmacytic lymphoma. Interestingly, loss of the Ink4a/Arf locus, apart from reducing the number of apoptotic B cells broadly attenuated KrasG12D -induced transcriptional signatures. However, combined Kras activation and Ink4a/Arf inactivation cooperated functionally to induce a fully penetrant, highly aggressive B-ALL phenotype resembling high-risk subtypes of human B-ALL such as BCR-ABL and CRFL2 -rearranged. Ninety percent of examined murine B-ALL tumors showed loss of the wild-type Ink4a/Arf locus without acquisition of highly recurrent cooperating events, underscoring the role of Ink4a/Arf in restraining Kras-driven oncogenesis in the lymphoid compartment. These data highlight the importance of functional cooperation between mutated Kras and Ink4a/Arf loss on B-ALL., Competing Interests: Conflict-of-interest disclosure: The authors declare no competing financial interests.- Published
- 2017
- Full Text
- View/download PDF
42. Subtyping sub-Saharan esophageal squamous cell carcinoma by comprehensive molecular analysis.
- Author
-
Liu W, Snell JM, Jeck WR, Hoadley KA, Wilkerson MD, Parker JS, Patel N, Mlombe YB, Mulima G, Liomba NG, Wolf LL, Shores CG, Gopal S, and Sharpless NE
- Published
- 2017
- Full Text
- View/download PDF
43. Lkb1 deletion in murine B lymphocytes promotes cell death and cancer.
- Author
-
Souroullas GP, Fedoriw Y, Staudt LM, and Sharpless NE
- Subjects
- AMP-Activated Protein Kinases, Animals, B-Lymphocytes pathology, Humans, Mice, Protein Serine-Threonine Kinases metabolism, T-Lymphocytes metabolism, T-Lymphocytes pathology, Apoptosis genetics, B-Lymphocytes metabolism, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, Cell Transformation, Neoplastic pathology, Gene Deletion, Lymphoma, B-Cell genetics, Lymphoma, B-Cell metabolism, Lymphoma, B-Cell pathology, Protein Serine-Threonine Kinases genetics
- Abstract
LKB1 (also known as STK11) is a potent tumor suppressor in solid tumors, such as melanoma and lung adenocarcinoma, but inactivation in hematopoietic cells causes cell death without signs of tumorigenesis. We noted somatic LKB1 deletion or mutation at low frequency in human B-cell lymphoma. To determine if LKB1 inactivation is a passenger or driver event in lymphoid cancers, we examined the effects of conditional inactivation of Lkb1 in murine lymphocytes. Consistent with prior reports, Lkb1 deletion in either T or B cells resulted in massive, lineage-specific apoptosis. Surprisingly, despite an 80% reduction of peripheral B-cell number, animals harboring somatic B-lineage Lkb1 deletion developed aggressive B-cell lymphoma with high penetrance and moderate latency. Malignant B cells exhibited somatic Lkb1 recombination. In contrast, Lkb1 deletion in T cells did not promote tumorigenesis. Concomitant Ras activation with Lkb1 deletion reduced T-cell apoptosis, but did not enhance tumor formation in T or B cells. These results suggest that although physiologic LKB1 expression exerts a potent pro-survival effect in lymphocytes, LKB1 inactivation nonetheless facilitates transformation of B, but not T, lymphocytes., (Copyright © 2017 ISEH - International Society for Experimental Hematology. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
44. Erratum: Lkb1 inactivation drives lung cancer lineage switching governed by Polycomb Repressive Complex 2.
- Author
-
Zhang H, Brainson CF, Koyama S, Redig AJ, Chen T, Li S, Gupta M, Garcia-de-Alba C, Paschini M, Herter-Sprie GS, Lu G, Zhang X, Marsh BP, Tuminello SJ, Xu C, Chen Z, Wang X, Akbay EA, Zheng M, Palakurthi S, Sholl LM, Rustgi AK, Kwiatkowski DJ, Alan Diehl J, Bass AJ, Sharpless NE, Dranoff G, Hammerman PS, Ji H, Bardeesy N, Saur D, Watanabe H, Kim CF, and Wong KK
- Abstract
This corrects the article DOI: 10.1038/ncomms14922.
- Published
- 2017
- Full Text
- View/download PDF
45. Senescence in Health and Disease.
- Author
-
He S and Sharpless NE
- Subjects
- Animals, Humans, Neoplasms immunology, Wound Healing, Aging pathology, Cell Cycle, Cellular Senescence
- Abstract
Many cellular stresses activate senescence, a persistent hyporeplicative state characterized in part by expression of the p16
INK4a cell-cycle inhibitor. Senescent cell production occurs throughout life and plays beneficial roles in a variety of physiological and pathological processes including embryogenesis, wound healing, host immunity, and tumor suppression. Meanwhile, the steady accumulation of senescent cells with age also has adverse consequences. These non-proliferating cells occupy key cellular niches and elaborate pro-inflammatory cytokines, contributing to aging-related diseases and morbidity. This model suggests that the abundance of senescent cells in vivo predicts "molecular," as opposed to chronologic, age and that senescent cell clearance may mitigate aging-associated pathology., (Copyright © 2017 Elsevier Inc. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
46. Transient CDK4/6 inhibition protects hematopoietic stem cells from chemotherapy-induced exhaustion.
- Author
-
He S, Roberts PJ, Sorrentino JA, Bisi JE, Storrie-White H, Tiessen RG, Makhuli KM, Wargin WA, Tadema H, van Hoogdalem EJ, Strum JC, Malik R, and Sharpless NE
- Subjects
- Animals, Bone Marrow Cells cytology, Bone Marrow Cells drug effects, Cell Cycle Checkpoints drug effects, Cell Proliferation drug effects, Cyclin-Dependent Kinase 4 antagonists & inhibitors, Cyclin-Dependent Kinase 4 metabolism, Cyclin-Dependent Kinase 6 antagonists & inhibitors, Cyclin-Dependent Kinase 6 metabolism, Female, Fluorouracil pharmacology, Healthy Volunteers, Hematopoietic Stem Cells cytology, Humans, Male, Mice, Mice, Inbred C57BL, Hematopoietic Stem Cells drug effects
- Abstract
Conventional cytotoxic chemotherapy is highly effective in certain cancers but causes dose-limiting damage to normal proliferating cells, especially hematopoietic stem and progenitor cells (HSPCs). Serial exposure to cytotoxics causes a long-term hematopoietic compromise ("exhaustion"), which limits the use of chemotherapy and success of cancer therapy. We show that the coadministration of G1T28 (trilaciclib), which is a small-molecule inhibitor of cyclin-dependent kinases 4 and 6 (CDK4/6), contemporaneously with cytotoxic chemotherapy protects murine hematopoietic stem cells (HSCs) from chemotherapy-induced exhaustion in a serial 5-fluorouracil treatment model. Consistent with a cell-intrinsic effect, we show directly preserved HSC function resulting in a more rapid recovery of peripheral blood counts, enhanced serial transplantation capacity, and reduced myeloid skewing. When administered to healthy human volunteers, G1T28 demonstrated excellent in vivo pharmacology and transiently inhibited bone marrow (BM) HSPC proliferation. These findings suggest that the combination of CDK4/6 inhibitors with cytotoxic chemotherapy should provide a means to attenuate therapy-induced BM exhaustion in patients with cancer., (Copyright © 2017, American Association for the Advancement of Science.)
- Published
- 2017
- Full Text
- View/download PDF
47. Lkb1 inactivation drives lung cancer lineage switching governed by Polycomb Repressive Complex 2.
- Author
-
Zhang H, Fillmore Brainson C, Koyama S, Redig AJ, Chen T, Li S, Gupta M, Garcia-de-Alba C, Paschini M, Herter-Sprie GS, Lu G, Zhang X, Marsh BP, Tuminello SJ, Xu C, Chen Z, Wang X, Akbay EA, Zheng M, Palakurthi S, Sholl LM, Rustgi AK, Kwiatkowski DJ, Diehl JA, Bass AJ, Sharpless NE, Dranoff G, Hammerman PS, Ji H, Bardeesy N, Saur D, Watanabe H, Kim CF, and Wong KK
- Subjects
- AMP-Activated Protein Kinases, Adenocarcinoma metabolism, Adenocarcinoma pathology, Animals, Carcinoma, Squamous Cell metabolism, Carcinoma, Squamous Cell pathology, Enhancer of Zeste Homolog 2 Protein genetics, Enhancer of Zeste Homolog 2 Protein metabolism, Gene Expression Profiling methods, Gene Expression Regulation, Neoplastic, Histones metabolism, Kaplan-Meier Estimate, Lung Neoplasms metabolism, Lung Neoplasms pathology, Methylation, Mice, 129 Strain, Mice, Knockout, Polycomb Repressive Complex 2 metabolism, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins p21(ras) genetics, Proto-Oncogene Proteins p21(ras) metabolism, Tumor Cells, Cultured, Adenocarcinoma genetics, Carcinoma, Squamous Cell genetics, Lung Neoplasms genetics, Polycomb Repressive Complex 2 genetics, Protein Serine-Threonine Kinases genetics
- Abstract
Adenosquamous lung tumours, which are extremely poor prognosis, may result from cellular plasticity. Here, we demonstrate lineage switching of KRAS+ lung adenocarcinomas (ADC) to squamous cell carcinoma (SCC) through deletion of Lkb1 (Stk11) in autochthonous and transplant models. Chromatin analysis reveals loss of H3K27me3 and gain of H3K27ac and H3K4me3 at squamous lineage genes, including Sox2, ΔNp63 and Ngfr. SCC lesions have higher levels of the H3K27 methyltransferase EZH2 than the ADC lesions, but there is a clear lack of the essential Polycomb Repressive Complex 2 (PRC2) subunit EED in the SCC lesions. The pattern of high EZH2, but low H3K27me3 mark, is also prevalent in human lung SCC and SCC regions within ADSCC tumours. Using FACS-isolated populations, we demonstrate that bronchioalveolar stem cells and club cells are the likely cells-of-origin for SCC transitioned tumours. These findings shed light on the epigenetics and cellular origins of lineage-specific lung tumours.
- Published
- 2017
- Full Text
- View/download PDF
48. Sustained p16 INK4a expression is required to prevent IR-induced tumorigenesis in mice.
- Author
-
Palacio L, Krishnan V, Le NL, Sharpless NE, and Beauséjour CM
- Subjects
- Animals, Cell Transformation, Neoplastic metabolism, Cell Transformation, Neoplastic radiation effects, Cyclin-Dependent Kinase Inhibitor p16 genetics, Female, Male, Mice, Mice, Knockout, Neoplasms, Experimental metabolism, Neoplasms, Experimental radiotherapy, Cell Transformation, Neoplastic pathology, Cyclin-Dependent Kinase Inhibitor p16 metabolism, Neoplasms, Experimental pathology, Radiation, Ionizing
- Abstract
Exposure of murine and human tissues to ionizing radiation (IR) induces the expression of p16
INK4a , a tumor suppressor gene and senescence/aging biomarker. Increased p16INK4a expression is often delayed several weeks post exposure to IR. In this context, it remains unclear if it occurs to suppress aberrant cellular growth of potentially transformed cells or is simply a result of IR-induced loss of tissue homeostasis. To address this question, we used a conditional p16INK4a null mouse model and determined the impact of p16INK4a inactivation long-term post exposure to IR. We found that, in vitro, bone marrow stromal cells exposed to IR enter DNA replication following p16INK4a inactivation. However, these cells did not resume growth; instead, they mostly underwent cell cycle arrest in G2. Similarly, delayed inactivation of p16INK4a in mice several weeks post exposure to IR resulted in increased BrdU incorporation and cancer incidence. In fact, we found that the onset of tumorigenesis was similar whether p16INK4a was inactivated before or after exposure to IR. Overall, our results suggest that IR-induced p16INK4a dependent growth arrest is reversible in mice and that sustained p16INK4a expression is necessary to protect against tumorigenesis.- Published
- 2017
- Full Text
- View/download PDF
49. p16 INK4a , a Senescence Marker, Influences Tenofovir/Emtricitabine Metabolite Disposition in HIV-Infected Subjects.
- Author
-
Dumond JB, Collins JW, Cottrell ML, Trezza CR, Prince H, Sykes C, Torrice C, White N, Malone S, Wang R, Patterson KB, Sharpless NE, and Forrest A
- Subjects
- Adult, Age Factors, Aged, Anti-HIV Agents administration & dosage, Cyclin-Dependent Kinase Inhibitor p16 genetics, Emtricitabine administration & dosage, Female, HIV Infections drug therapy, HIV Infections genetics, Humans, Male, Middle Aged, Nonlinear Dynamics, Tenofovir pharmacology, Young Adult, Anti-HIV Agents pharmacokinetics, Cyclin-Dependent Kinase Inhibitor p16 metabolism, Emtricitabine pharmacokinetics, HIV Infections metabolism, Tenofovir administration & dosage
- Abstract
The goal of this study was to explore the relationships between tenofovir (TFV) and emtricitabine (FTC) disposition and markers of biologic aging, such as the frailty phenotype and p16
INK4a gene expression. Chronologic age is often explored in population pharmacokinetic (PK) analyses, and can be uninformative in capturing the impact of aging on physiology, particularly in human immunodeficiency virus (HIV)-infected patients. Ninety-one HIV-infected participants provided samples to quantify plasma concentrations of TFV/FTC, as well as peripheral blood mononuclear cell (PBMC) samples for intracellular metabolite concentrations; 12 participants provided 11 samples, and 79 participants provided 4 samples, over a dosing interval. Nonlinear mixed effects modeling of TFV/FTC and their metabolites suggests a relationship between TFV/FTC metabolite clearance (CL) from PBMCs and the expression of p16INK4a , a marker of cellular senescence. This novel approach to quantifying the influence of aging on PKs provides rationale for further work investigating the relationships between senescence and nucleoside phosphorylation and transport., (© 2016 The Authors CPT: Pharmacometrics & Systems Pharmacology published by Wiley Periodicals, Inc. on behalf of American Society for Clinical Pharmacology and Therapeutics.)- Published
- 2017
- Full Text
- View/download PDF
50. Cellular Senescence Promotes Adverse Effects of Chemotherapy and Cancer Relapse.
- Author
-
Demaria M, O'Leary MN, Chang J, Shao L, Liu S, Alimirah F, Koenig K, Le C, Mitin N, Deal AM, Alston S, Academia EC, Kilmarx S, Valdovinos A, Wang B, de Bruin A, Kennedy BK, Melov S, Zhou D, Sharpless NE, Muss H, and Campisi J
- Subjects
- Animals, Antineoplastic Agents pharmacology, Breast Neoplasms genetics, Breast Neoplasms metabolism, Breast Neoplasms pathology, Cell Proliferation drug effects, Cyclin-Dependent Kinase Inhibitor p16 metabolism, Female, Humans, Mice, Mice, Transgenic, Neoplasm Recurrence, Local, Antineoplastic Agents adverse effects, Breast Neoplasms drug therapy, Cellular Senescence, Cyclin-Dependent Kinase Inhibitor p16 genetics
- Abstract
Cellular senescence suppresses cancer by irreversibly arresting cell proliferation. Senescent cells acquire a proinflammatory senescence-associated secretory phenotype. Many genotoxic chemotherapies target proliferating cells nonspecifically, often with adverse reactions. In accord with prior work, we show that several chemotherapeutic drugs induce senescence of primary murine and human cells. Using a transgenic mouse that permits tracking and eliminating senescent cells, we show that therapy-induced senescent (TIS) cells persist and contribute to local and systemic inflammation. Eliminating TIS cells reduced several short- and long-term effects of the drugs, including bone marrow suppression, cardiac dysfunction, cancer recurrence, and physical activity and strength. Consistent with our findings in mice, the risk of chemotherapy-induced fatigue was significantly greater in humans with increased expression of a senescence marker in T cells prior to chemotherapy. These findings suggest that senescent cells can cause certain chemotherapy side effects, providing a new target to reduce the toxicity of anticancer treatments., Significance: Many genotoxic chemotherapies have debilitating side effects and also induce cellular senescence in normal tissues. The senescent cells remain chronically present where they can promote local and systemic inflammation that causes or exacerbates many side effects of the chemotherapy. Cancer Discov; 7(2); 165-76. ©2016 AACR.This article is highlighted in the In This Issue feature, p. 115., Competing Interests: All other authors declare no financial interests., (©2016 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.