Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Peter S. Kim is active.

Publication


Featured researches published by Peter S. Kim.


The Journal of Infectious Diseases | 2012

Tuberculosis Diagnostics and Biomarkers: Needs, Challenges, Recent Advances, and Opportunities

Ruth McNerney; Markus Maeurer; Ibrahim Abubakar; Ben J. Marais; Timothy D. McHugh; Nathan Ford; Karin Weyer; Steve Lawn; Martin P. Grobusch; Ziad A. Memish; S. Bertel Squire; Giuseppe Pantaleo; Jeremiah Chakaya; Martina Casenghi; Giovanni Batista Migliori; Peter Mwaba; Lynn S. Zijenah; Michael Hoelscher; Helen Cox; Soumya Swaminathan; Peter S. Kim; Marco Schito; Alexandre Harari; Matthew Bates; Samana Schwank; Justin O'Grady; Michel Pletschette; Lucica Ditui; Rifat Atun; Alimuddin Zumla

Tuberculosis is unique among the major infectious diseases in that it lacks accurate rapid point-of-care diagnostic tests. Failure to control the spread of tuberculosis is largely due to our inability to detect and treat all infectious cases of pulmonary tuberculosis in a timely fashion, allowing continued Mycobacterium tuberculosis transmission within communities. Currently recommended gold-standard diagnostic tests for tuberculosis are laboratory based, and multiple investigations may be necessary over a period of weeks or months before a diagnosis is made. Several new diagnostic tests have recently become available for detecting active tuberculosis disease, screening for latent M. tuberculosis infection, and identifying drug-resistant strains of M. tuberculosis. However, progress toward a robust point-of-care test has been limited, and novel biomarker discovery remains challenging. In the absence of effective prevention strategies, high rates of early case detection and subsequent cure are required for global tuberculosis control. Early case detection is dependent on test accuracy, accessibility, cost, and complexity, but also depends on the political will and funder investment to deliver optimal, sustainable care to those worst affected by the tuberculosis and human immunodeficiency virus epidemics. This review highlights unanswered questions, challenges, recent advances, unresolved operational and technical issues, needs, and opportunities related to tuberculosis diagnostics.


AIDS | 2011

Efficacy of a nucleoside-sparing regimen of darunavir/ritonavir plus raltegravir in treatment-naive HIV-1-infected patients (ACTG A5262).

Babafemi Taiwo; Lu Zheng; Sébastien Gallien; Roy M. Matining; Daniel R. Kuritzkes; Cara C. Wilson; Baiba Berzins; Edward P. Acosta; Barbara Bastow; Peter S. Kim; Joseph J. Eron

Objective:To explore darunavir/ritonavir (DRV/r) plus raltegravir (RAL) combination therapy in antiretroviral-naive patients. Design:Phase IIb, single-arm, open-label, multicenter study. Methods:One hundred and twelve antiretroviral-naive, HIV-1-infected patients received DRV/r 800/100u200amg once daily and RAL 400u200amg twice daily. Primary endpoint was virologic failure by week 24. Virologic failure was defined as confirmed viral load of 1000u200acopies/ml or more at week 12, or an increase of more than 0.5u200alog10u200acopies/ml in viral load from week 4 to 12, or a confirmed viral load of more than 50u200acopies/ml at or after week 24. Protease and integrase genes were sequenced in patients experiencing virologic failure. Results:Virologic failure rate was 16% [95% confidence interval (CI) 10–24] by week 24 and 26% (95% CI 19–36) by week 48 in an intent-to-treat analysis. Viral load at virologic failure was 51–200u200acopies/ml in 17/28 failures. Adjusting for age and sex, virologic failure was associated with baseline viral load of more than 100u200a000u200acopies/ml [hazard ratio 3.76, 95% CI (1.52–9.31), Pu200a=u200a0.004] and lower CD4 cell count [0.77 per 100u200acells/&mgr;l increase (95% CI 0.61–0.98), Pu200a=u200a0.037]. When trough RAL concentrations were included as a time-varying covariate in the analysis, virologic failure remained associated with baseline viral load more than 100u200a000u200acopies/ml [hazard ratiou200a=u200a4.67 (95% CI 1.93–11.25), Pu200a<u200a0.001], whereas RAL level below detection limit in plasma at one or more previous visits was associated with increased hazard [hazard ratiou200a=u200a3.42 (95% CI 1.41–8.26), Pu200a=u200a0.006]. All five participants with integrase mutations during virologic failure had baseline viral load more than 100u200a000u200acopies/ml. Conclusion:DRV/r plus RAL was effective and well tolerated in most patients, but virologic failure and integrase resistance were common, particularly in patients with baseline viral load more than 100u200a000u200acopies/ml.


Diabetes Care | 2009

A1C Underestimates Glycemia in HIV Infection

Peter S. Kim; Christian Woods; Patrick Georgoff; Dana Crum; Alice Rosenberg; Margo A. Smith; Colleen Hadigan

OBJECTIVE The objective of this study was to determine the relationship between A1C and glycemia in HIV infection. RESEARCH DESIGN AND METHODS We completed a prospective cross-sectional study of 100 HIV-infected adults with type 2 diabetes (77%) or fasting hyperglycemia (23%) with measured glucose, A1C, mean corpuscular volume (MCV), and fructosamine. A total of 200 HIV-uninfected type 2 diabetic subjects matched for key demographic characteristics served as control subjects. RESULTS Relative to the control subjects, A1C underestimated glucose by 29 ± 4 mg/dl in the HIV-infected subjects. Current nucleoside reverse transcriptase inhibitors (NRTIs), higher MCV and hemoglobin, and lower HIV RNA and haptoglobin were associated with greater A1C-glucose discordance. However, only MCV and current NTRI use, in particular abacavir, remained significant predictors in multivariate analyses. Fructosamine more closely reflected glycemia in the HIV-infected subjects. CONCLUSIONS A1C underestimates glycemia in HIV-infected patients and is related to NRTI use. Use of abacavir and increased MCV were key correlates in multivariate analyses. Fructosamine may be more appropriate in this setting.


Oncotarget | 2016

IL-15 superagonist/IL-15RαSushi-Fc fusion complex (IL-15SA/IL-15RαSu-Fc; ALT-803) markedly enhances specific subpopulations of NK and memory CD8+ T cells, and mediates potent anti-tumor activity against murine breast and colon carcinomas

Peter S. Kim; Anna R. Kwilas; Wenxin Xu; Sarah Alter; Emily K. Jeng; Hing C. Wong; Jeffrey Schlom; James W. Hodge

Interleukin (IL)-15-N72D superagonist-complexed with IL-15RαSushi-Fc fusion protein (IL-15SA/IL-15RαSu-Fc; ALT-803) has been reported to exhibit significant anti-tumor activity in murine myeloma, rat bladder cancer, and murine glioblastoma models. In this study, we examined the immunomodulatory and anti-tumor effects of IL-15SA/IL-15RαSu-Fc in tumor-free and highly metastatic tumor-bearing mice. Here, IL-15SA/IL-15RαSu-Fc significantly expanded natural killer (NK) and CD8+ T cells. In examining NK cell subsets, the greatest significant increase was in highly cytotoxic and migrating (CD11b+, CD27hi; high effector) NK cells, leading to enhanced function on a per-cell basis. CD8+ T cell subset analysis determined that IL-15SA/IL-15RαSu-Fc significantly increased IL-15 responding memory (CD122+, CD44+) CD8+ T cells, in particular those having the innate (NKG2D+, PD1−) phenotype. In 4T1 breast tumor–bearing mice, IL-15SA/IL-15RαSu-Fc induced significant anti-tumor activity against spontaneous pulmonary metastases, depending on CD8+ T and NK cells, and resulting in prolonged survival. Similar anti-tumor activity was seen in the experimental pulmonary metastasis model of CT26 colon carcinoma cells, particularly when IL-15SA/IL-15RαSu-Fc was combined with a cocktail of checkpoint inhibitors, anti-CTLA-4 and anti-PD-L1. Altogether, these studies showed for the first time that IL-15SA/IL-15RαSu-Fc (1) promoted the development of high effector NK cells and CD8+ T cell responders of the innate phenotype, (2) enhanced function of NK cells, and (3) played a vital role in reducing tumor metastasis and ultimately survival, especially in combination with checkpoint inhibitors.


European Respiratory Journal | 2017

A standardised method for interpreting the association between mutations and phenotypic drug resistance in Mycobacterium tuberculosis

Paolo Miotto; Belay Tessema; Elisa Tagliani; Leonid Chindelevitch; Angela M. Starks; Claudia Emerson; Debra Hanna; Peter S. Kim; Richard Liwski; Matteo Zignol; Christopher Gilpin; Stefan Niemann; Claudia M. Denkinger; Joy Fleming; Robin M. Warren; Derrick W. Crook; James E. Posey; Sebastien Gagneux; Sven Hoffner; Camilla Rodrigues; Iñaki Comas; David M. Engelthaler; Megan Murray; David Alland; Leen Rigouts; Christoph Lange; Keertan Dheda; Rumina Hasan; Uma Devi Ranganathan; Ruth McNerney

A clear understanding of the genetic basis of antibiotic resistance in Mycobacterium tuberculosis is required to accelerate the development of rapid drug susceptibility testing methods based on genetic sequence. Raw genotype–phenotype correlation data were extracted as part of a comprehensive systematic review to develop a standardised analytical approach for interpreting resistance associated mutations for rifampicin, isoniazid, ofloxacin/levofloxacin, moxifloxacin, amikacin, kanamycin, capreomycin, streptomycin, ethionamide/prothionamide and pyrazinamide. Mutation frequencies in resistant and susceptible isolates were calculated, together with novel statistical measures to classify mutations as high, moderate, minimal or indeterminate confidence for predicting resistance. We identified 286 confidence-graded mutations associated with resistance. Compared to phenotypic methods, sensitivity (95% CI) for rifampicin was 90.3% (89.6–90.9%), while for isoniazid it was 78.2% (77.4–79.0%) and their specificities were 96.3% (95.7–96.8%) and 94.4% (93.1–95.5%), respectively. For second-line drugs, sensitivity varied from 67.4% (64.1–70.6%) for capreomycin to 88.2% (85.1–90.9%) for moxifloxacin, with specificity ranging from 90.0% (87.1–92.5%) for moxifloxacin to 99.5% (99.0–99.8%) for amikacin. This study provides a standardised and comprehensive approach for the interpretation of mutations as predictors of M. tuberculosis drug-resistant phenotypes. These data have implications for the clinical interpretation of molecular diagnostics and next-generation sequencing as well as efficient individualised therapy for patients with drug-resistant tuberculosis. A comprehensive basis for interpreting mutations to predict antibiotic resistance in tuberculosis http://ow.ly/hhwJ30g9jCY


Clinical Infectious Diseases | 2015

Collaborative Effort for a Centralized Worldwide Tuberculosis Relational Sequencing Data Platform

Angela M. Starks; Enrique Aviles; Daniela M. Cirillo; Claudia M. Denkinger; David L. Dolinger; Claudia Emerson; Jim Gallarda; Debra Hanna; Peter S. Kim; Richard Liwski; Paolo Miotto; Marco Schito; Matteo Zignol

Continued progress in addressing challenges associated with detection and management of tuberculosis requires new diagnostic tools. These tools must be able to provide rapid and accurate information for detecting resistance to guide selection of the treatment regimen for each patient. To achieve this goal, globally representative genotypic, phenotypic, and clinical data are needed in a standardized and curated data platform. A global partnership of academic institutions, public health agencies, and nongovernmental organizations has been established to develop a tuberculosis relational sequencing data platform (ReSeqTB) that seeks to increase understanding of the genetic basis of resistance by correlating molecular data with results from drug susceptibility testing and, optimally, associated patient outcomes. These data will inform development of new diagnostics, facilitate clinical decision making, and improve surveillance for drug resistance. ReSeqTB offers an opportunity for collaboration to achieve improved patient outcomes and to advance efforts to prevent and control this devastating disease.


The Journal of Infectious Diseases | 2012

Clinical research and development of tuberculosis diagnostics: moving from silos to synergy.

Payam Nahid; Peter S. Kim; Carlton A. Evans; David Alland; Michael R. Barer; Jane Diefenbach; Jerrold J. Ellner; Richard Hafner; Carol Hamilton; Michael F. Iademarco; Gregory C. Ireton; Michael E. Kimerling; Christian Lienhardt; William R. MacKenzie; Megan Murray; Mark D. Perkins; Jamie E. Posey; Teri Roberts; Christine Sizemore; Wendy Stevens; Laura E. Via; Sharon D. Williams; Wing Yew; Susan Swindells

The development, evaluation, and implementation of new and improved diagnostics have been identified as critical needs by human immunodeficiency virus (HIV) and tuberculosis researchers and clinicians alike. These needs exist in international and domestic settings and in adult and pediatric populations. Experts in tuberculosis and HIV care, researchers, healthcare providers, public health experts, and industry representatives, as well as representatives of pertinent US federal agencies (Centers for Disease Control and Prevention, Food and Drug Administration, National Institutes of Health, United States Agency for International Development) assembled at a workshop proposed by the Diagnostics Working Group of the Federal Tuberculosis Taskforce to review the state of tuberculosis diagnostics development in adult and pediatric populations.


Aids Research and Treatment | 2012

TB and HIV Therapeutics: Pharmacology Research Priorities

Kelly E. Dooley; Peter S. Kim; Sharon D. Williams; Richard Hafner

An unprecedented number of investigational drugs are in the development pipeline for the treatment of tuberculosis. Among patients with tuberculosis, co-infection with HIV is common, and concurrent treatment of tuberculosis and HIV is now the standard of care. To ensure that combinations of anti-tuberculosis drugs and antiretrovirals are safe and are tested at doses most likely to be effective, selected pharmacokinetic studies based on knowledge of their metabolic pathways and their capacity to induce or inhibit metabolizing enzymes of companion drugs must be conducted. Drug interaction studies should be followed up by evaluations in larger populations to evaluate safety and pharmacodynamics more fully. Involving patients with HIV in trials of TB drugs early in development enhances the knowledge gained from the trials and will ensure that promising new tuberculosis treatments are available to patients with HIV as early as possible. In this review, we summarize current and planned pharmacokinetic and drug interaction studies involving investigational and licensed tuberculosis drugs and antiretrovirals and suggest priorities for tuberculosis-HIV pharmacokinetic, pharmacodynamic, and drug-drug interaction studies for the future. Priority studies for children and pregnant women with HIV and tuberculosis co-infection are briefly discussed.


PLOS ONE | 2011

Increased prevalence of albuminuria in HIV-infected adults with diabetes.

Peter S. Kim; Christian Woods; Lauren Dutcher; Patrick Georgoff; Alice Rosenberg; Jo Ann M. Mican; Jeffrey B. Kopp; Margo A. Smith; Colleen Hadigan

Objective HIV and type 2 diabetes are known risk factors for albuminuria, but no previous reports have characterized albuminuria in HIV-infected patients with diabetes. Research Design and Methods We performed a cross-sectional study including 73 HIV-infected adults with type 2 diabetes, 82 HIV-infected non-diabetics, and 61 diabetic control subjects without HIV. Serum creatinine >1.5 mg/dL was exclusionary. Albuminuria was defined as urinary albumin/creatinine ratio >30 mg/g. Results The prevalence of albuminuria was significantly increased among HIV-infected diabetics (34% vs. 13% of HIV non-diabetic vs. 16% diabetic control, pu200a=u200a0.005). HIV status and diabetes remained significant predictors of albuminuria after adjusting for age, race, BMI, and blood pressure. Albumin/creatinine ratio correlated significantly with HIV viral load (ru200a=u200a0.28, pu200a=u200a0.0005) and HIV-infected subjects with albuminuria had significantly greater cumulative exposure to abacavir (pu200a=u200a0.01). In an adjusted multivariate regression analysis of HIV-infected subjects, the diagnosis of diabetes (pu200a=u200a0.003), higher HIV viral load (pu200a=u200a0.03) and cumulative exposure to abacavir (pu200a=u200a0.0009) were significant independent predictors of albuminuria. Conclusions HIV and diabetes appear to have additive effects on albuminuria which is also independently associated with increased exposure to abacavir and HIV viral load. Future research on the persistence, progression and management of albuminuria in this unique at-risk population is needed.


Wiley Interdisciplinary Reviews-nanomedicine and Nanobiotechnology | 2010

Nanotechnology and HIV: potential applications for treatment and prevention

Peter S. Kim; Sarah W. Read

HIV/AIDS is a global pandemic and is the leading infectious cause of death among adults. Although antiretroviral (ARV) therapy has dramatically improved the quality of life and increased the life expectancy of those infected with HIV, life-long suppressive treatment is required and a cure for HIV infection remains elusive; frequency of dosing and drug toxicity as well as the development of viral resistance pose additional limitations. Furthermore, preventative measures such as a vaccine or microbicide are urgently needed to curb the rate of new infections. The capabilities inherent to nanotechnology hold much potential for impact in the field of HIV treatment and prevention. This article reviews the potential for the multidisciplinary field of nanotechnology to advance the fields of HIV treatment and prevention.

Collaboration


Dive into the Peter S. Kim's collaboration.

Top Co-Authors

Avatar

Jeffrey Schlom

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Anna R. Kwilas

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Benedetto Farsaci

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Italia Grenga

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

James W. Hodge

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Renee N. Donahue

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Richard Hafner

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

James L. Gulley

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Caroline Jochems

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge