全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Point of Care Technologies for HIV

DOI: 10.1155/2014/497046

Full-Text   Cite this paper   Add to My Lib

Abstract:

Effective prevention of HIV/AIDS requires early diagnosis, initiation of therapy, and regular plasma viral load monitoring of the infected individual. In addition, incidence estimation using accurate and sensitive assays is needed to facilitate HIV prevention efforts in the public health setting. Therefore, more affordable and accessible point-of-care (POC) technologies capable of providing early diagnosis, HIV viral load measurements, and CD4 counts in settings where HIV is most prevalent are needed to enable appropriate intervention strategies and ultimately stop transmission of the virus within these populations to achieve the future goal of an AIDS-free generation. This review discusses the available and emerging POC technologies for future application to these unmet public health needs. 1. Introduction Acquired immunodeficiency syndrome (AIDS) is a disease of the human immune system caused by the HIV [1] which destroys CD4+ T lymphocytes of the immune system that prevent infections. AIDS is one of the most serious global health problems of unprecedented dimensions and is one of the greatest modern pandemics. At the end of 2010, an estimated 34 million people were living with HIV globally, including 3.4 million children under 15 years of age. There were 2.7 million new HIV infections in 2010, including 390?000 among children less than 15 years [2]. The annual number of people newly infected with the HIV has declined 20% from the global epidemic peak in 1998, mainly due to tremendous progress in diagnostics and treatment. Several diagnostic technologies have emerged with high specificity, sensitivity, and accuracy to detect HIV infection. HIV testing plays an important role in HIV prevention in that knowledge of HIV status has both individual and public health benefits. Early and accurate detection of HIV infection is important to public health because this stage is characterized by high infectiousness and transmissibility of the virus. The individual benefits of HIV testing are primarily associated with individuals accessing care and treatment. Individuals entering care and treatment have a substantial reduction in adverse health outcomes and increased life expectancy. The past decades have witnessed enormous technological improvements towards the development of simple, cost-effective, and accurate rapid diagnostic tests for detection and identification of infectious pathogens. There is growing demand within the global health community to find ways to simplify and improve the efficiency of diagnostics for HIV/AIDS without diminishing the quality of

References

[1]  K. A. S. Pkowitz, “AIDS—the first 20 years,” The New England Journal of Medicine, vol. 344, no. 23, pp. 1764–1772, 2001.
[2]  WHO, UNICEF, and UNAIDS, “Epidemic update and health sector progress towards universal access,” Progress Report: Global HIV/AIDS Response, WHO, Geneva, Switzerland, 2011.
[3]  UNITAID, “HIV/AIDS diagnostic technology landscape,” Tech. Rep., WHO, Geneva, Switzerland, 2012.
[4]  R. J. Meagher, A. V. Hatch, R. F. Renzi, and A. K. Singh, “An integrated microfluidic platform for sensitive and rapid detection of biological toxins,” Lab on a Chip, vol. 8, no. 12, pp. 2046–2053, 2008.
[5]  J. Wang, H. Ahmad, C. Ma et al., “A self-powered, one-step chip for rapid, quantitative and multiplexed detection of proteins from pinpricks of whole blood,” Lab on a Chip, vol. 10, no. 22, pp. 3157–3162, 2010.
[6]  C. J?nsson, M. Aronsson, G. Rundstr?m et al., “Silane-dextran chemistry on lateral flow polymer chips for immunoassays,” Lab on a Chip, vol. 8, no. 7, pp. 1191–1197, 2008.
[7]  M. M. Murtagh and T. F. Peter, “ART 2.0: implications for diagnostics in resource-limited settings,” Clinton Health Access Initiative, 2010.
[8]  G. Wu and M. H. Zaman, “Low-cost tools for diagnosing and monitoring HIV infection in low-resource settings,” Bulletin of the WHO, vol. 90, pp. 914–920, 2012.
[9]  M. R. Parisi, L. Soldini, G. D. Perri, S. Tiberi, A. Lazzarin, and F. B. Lillo, “Offer of rapid testing and alternative biological samples as practical tools to implement HIV screening programs,” New Microbiologica, vol. 32, no. 4, pp. 391–396, 2009.
[10]  B. M. Branson, “The future of HIV testing,” Journal of Acquired Immune Deficiency Syndromes, vol. 55, supplement 2, pp. S102–S105, 2010.
[11]  G. Beelaert and K. Fransen, “Evaluation of a rapid and simple fourth-generation HIV screening assay for qualitative detection of HIV p24 antigen and/or antibodies to HIV-1 and HIV-2,” Journal of Virological Methods, vol. 168, no. 1-2, pp. 218–222, 2010.
[12]  J. Fox, H. Dunn, and S. O'Shea, “Low rates of p24 antigen detection using a fourth-generation point of care HIV test,” Sexually Transmitted Infections, vol. 87, no. 2, pp. 178–179, 2011.
[13]  S. Sivapalasingam, S. Essajee, P. N. Nyambi et al., “Human immunodeficiency virus (HIV) reverse transcriptase activity correlates with HIV RNA load: implications for resource-limited settings,” Journal of Clinical Microbiology, vol. 43, no. 8, pp. 3793–3796, 2005.
[14]  K. Steegen, S. Luchters, N. De Cabooter et al., “Evaluation of two commercially available alternatives for HIV-1 viral load testing in resource-limited settings,” Journal of Virological Methods, vol. 146, no. 1-2, pp. 178–187, 2007.
[15]  S. Sivapalasingam, B. Wangechi, F. Marshed et al., “Monitoring virologic responses to antiretroviral therapy in HIV-infected adults in Kenya: evaluation of a low-cost viral load assay,” PLoS One, vol. 4, no. 8, Article ID e6828, 2009.
[16]  W. Labbett, A. Garcia-Diaz, Z. Fox et al., “Comparative evaluation of the ExaVir load version 3 reverse transcriptase assay for measurement of human immunodeficiency virus type 1 plasma load,” Journal of Clinical Microbiology, vol. 47, no. 10, pp. 3266–3270, 2009.
[17]  D. Bonard, F. Rouet, T. A. Toni et al., “Field evaluation of an improved assay using a heat-dissociated p24 antigen for adults mainly infected with HIV-1 CRF02_AG strains in C?te d'Ivoire, West Africa,” Journal of Acquired Immune Deficiency Syndromes, vol. 34, no. 3, pp. 267–273, 2003.
[18]  V. L. Greengrass, M. M. Plate, P. M. Steele et al., “Evaluation of the Cavidi ExaVir Load assay (version 3) for plasma human immunodeficiency virus type 1 load monitoring,” Journal of Clinical Microbiology, vol. 47, no. 9, pp. 3011–3013, 2009.
[19]  R. Zachariah, S. D. Reid, P. Chaillet, M. Massaquoi, E. J. Schouten, and A. D. Harries, “Viewpoint: why do we need a point-of-care CD4 test for low-income countries?” Tropical Medicine and International Health, vol. 16, no. 1, pp. 37–41, 2011.
[20]  W. H. A. Chow, C. McCloskey, Y. Tong et al., “Application of isothermal helicase-dependent amplification with a disposable detection device in a simple sensitive stool test for toxigenic Clostridium difficile,” Journal of Molecular Diagnostics, vol. 10, no. 5, pp. 452–458, 2008.
[21]  T. D. Ly, S. Laperche, C. Brennan et al., “Evaluation of the sensitivity and specificity of six HIV combined p24 antigen and antibody assays,” Journal of Virological Methods, vol. 122, no. 2, pp. 185–194, 2004.
[22]  T. D. Ly, L. Martin, D. Daghfal et al., “Seven human immunodeficiency virus (HIV) antigen-antibody combination assays: evaluation of HIV seroconversion sensitivity and subtype detection,” Journal of Clinical Microbiology, vol. 39, no. 9, pp. 3122–3128, 2001.
[23]  G. Murphy and C. Aitken, “HIV testing-the perspective from across the pond,” Journal of Clinical Virology, vol. 52, supplement 1, pp. S71–S76, 2011.
[24]  B. Weber, “Screening of HIV infection: role of molecular and immunological assays,” Expert Review of Molecular Diagnostics, vol. 6, no. 3, pp. 399–411, 2006.
[25]  B. Weber, L. Gürtler, R. Thorstensson et al., “Multicenter evaluation of a new automated fourth-generation human immunodeficiency virus screening assay with a sensitive antigen detection module and high specificity,” Journal of Clinical Microbiology, vol. 40, no. 6, pp. 1938–1946, 2002.
[26]  T. D. Ly, A. Ebel, V. Faucher, V. Fihman, and S. Laperche, “Could the new HIV combined p24 antigen and antibody assays replace p24 antigen specific assays?” Journal of Virological Methods, vol. 143, no. 1, pp. 86–94, 2007.
[27]  S. P. Layne, M. J. Merges, M. Dembo et al., “Factors underlying spontaneous inactivation and susceptibility to neutralization of human immunodeficiency virus,” Virology, vol. 189, no. 2, pp. 695–714, 1992.
[28]  C. Bentsen, L. McLaughlin, E. Mitchell et al., “Performance evaluation of the Bio-Rad Laboratories GS HIV Combo Ag/Ab EIA, a 4th generation HIV assay for the simultaneous detection of HIV p24 antigen and antibodies to HIV-1 (groups M and O) and HIV-2 in human serum or plasma,” Journal of Clinical Virology, vol. 52, supplement 1, pp. S57–S61, 2011.
[29]  P. Chavez, L. Wesolowski, P. Patel, K. Delaney, and S. M. Owen, “Evaluation of the performance of the Abbott ARCHITECT HIV Ag/Ab combo assay,” Journal of Clinical Virology, vol. 52, supplement 1, pp. S51–S55, 2011.
[30]  M. W. Pandori, J. Hackett Jr., B. Louie et al., “Assessment of the ability of a fourth-generation immunoassay for human immunodeficiency virus (HIV) antibody and p24 antigen to detect both acute and recent HIV infections in a high-risk setting,” Journal of Clinical Microbiology, vol. 47, no. 8, pp. 2639–2642, 2009.
[31]  L. Laursen, “Point-of-care tests poised to alter course of HIV treatment,” Nature Medicine, vol. 18, article 1156, 2012.
[32]  A. Cachafeiro, G. G. Sherman, A. H. Sohn, C. Beck-Sague, and S. A. Fiscus, “Diagnosis of human immunodeficiency virus type 1 infection in infants by use of dried blood spots and an ultrasensitive p24 antigen assay,” Journal of Clinical Microbiology, vol. 47, no. 2, pp. 459–462, 2009.
[33]  S. Workman, S. K. Wells, C.-P. Pau et al., “Rapid detection of HIV-1 p24 antigen using magnetic immuno-chromatography (MICT),” Journal of Virological Methods, vol. 160, no. 1-2, pp. 14–21, 2009.
[34]  Z. A. Parpia, R. Elghanian, A. Nabatiyan, D. R. Hardie, and D. M. Kelso, “P24 antigen rapid test for diagnosis of acute pediatric HIV infection,” Journal of Acquired Immune Deficiency Syndromes, vol. 55, no. 4, pp. 413–419, 2010.
[35]  M. M. Murtagh, “HIV/AIDS diagnostics landscape,” UNITAID Technical Report, WHO, Geneva, Switzerland, 2011.
[36]  S. Mtapuri-Zinyowera, M. Chideme, D. Mangwanya et al., “Evaluation of the PIMA point-of-care CD4 analyzer in VCT clinics in Zimbabwe,” Journal of Acquired Immune Deficiency Syndromes, vol. 55, no. 1, pp. 1–7, 2010.
[37]  N. Sitoe, E. Luecke, N. Tembe et al., “Absolute and percent CD4+ T-cell enumeration by flow cytometry using capillary blood,” Journal of Immunological Methods, vol. 372, no. 1-2, pp. 1–6, 2011.
[38]  Y. C. Manabe, Y. Wang, A. Elbireer, B. Auerbach, and B. Castelnuovo, “Evaluation of portable point-of-care CD4 counter with high sensitivity for detecting patients eligible for antiretroviral therapy,” PLoS One, vol. 7, no. 4, Article ID e34319, 2012.
[39]  D. K. Glencross, L. M. Coetzee, M. Faal et al., “Performance evaluation of the Pima point-of-care CD4 analyser using capillary blood sampling in field tests in South Africa,” Journal of the International AIDS Society, vol. 15, no. 1, article 3, 2012.
[40]  S. Tanriverdi, L. Chen, and S. Chen, “A rapid and automated sample-to-result HIV load test for near-patient application,” The Journal of Infectious Diseases, vol. 201, supplement 1, pp. S52–S58, 2010.
[41]  R. L. Hodinka, T. Nagashunmugam, and D. Malamud, “Detection of human immunodeficiency virus antibodies in oral fluids,” Clinical and Diagnostic Laboratory Immunology, vol. 5, no. 4, pp. 419–426, 1998.
[42]  W. A. O'Brien, P. M. Hartigan, D. Martin et al., “Changes in plasma HIV-1 RNA and CD4+ lymphocyte counts and the risk of progression to AIDS,” The New England Journal of Medicine, vol. 334, no. 7, pp. 426–431, 1996.
[43]  J. A. Sterne, M. May, D. Costagliola et al., “Timing of initiation of antiretroviral therapy in AIDS-free HIV-1-infected patients: a collaborative analysis of 18 HIV cohort studies,” The Lancet, vol. 373, no. 9672, pp. 1352–1363, 2009.
[44]  WHO, “Antiretroviral therapy for HIV infection in adults and adolescents, recommendations for a public health approach,” 2010 Revision, WHO, Geneva, Switzerland, 2010.
[45]  “Review of CD4 technologies,” G.S. Presentation, Geneva: World Health Organization, 2011, http://www.who.int/hiv/topics/treatment.
[46]  I. V. Jani, N. E. Sitoe, P. L. Chongo et al., “Accurate CD4 T-cell enumeration and antiretroviral drug toxicity monitoring in primary healthcare clinics using point-of-care testing,” Journal of the International AIDS Society, vol. 25, no. 6, pp. 807–812, 2011.
[47]  X. Cheng, D. Irimia, M. Dixon et al., “A microchip approach for practical label-free CD4+ T-cell counting of HIV-infected subjects in resource-poor settings,” Journal of Acquired Immune Deficiency Syndromes, vol. 45, no. 3, pp. 257–261, 2007.
[48]  X. Cheng, Y.-S. Liu, D. Irimia et al., “Cell detection and counting through cell lysate impedance spectroscopy in microfluidic devices,” Lab on a Chip, vol. 7, no. 6, pp. 746–755, 2007.
[49]  D. S. Boyle, K. R. Hawkins, M. S. Steele, M. Singhal, and X. Cheng, “Emerging technologies for point-of-care CD4 T-lymphocyte counting,” Trends in Biotechnology, vol. 30, no. 1, pp. 45–54, 2012.
[50]  C. Willyard, “Simpler tests for immune cells could transform AIDS care in Africa,” Nature Medicine, vol. 13, no. 10, p. 1131, 2007.
[51]  D. L. Robertson, J. P. Anderson, J. A. Bradac et al., “HIV-1 nomenclature proposal,” Science, vol. 288, no. 5463, pp. 55–56, 2000.
[52]  “HIV sequence database,” 2012, http://www.hiv.lanl.gov/content/sequence/HIV/CRFs/CRFs.html.
[53]  WHO, UNICEF, and UNAIDS, “Towards universal access: scaling up priority HIV/AIDS interventions in the health sector,” Progress Report, WHO, Geneva, Switzerland, 2009.
[54]  S. Wang, F. Xu, and U. Demirci, “Advances in developing HIV-1 viral load assays for resource-limited settings,” Biotechnology Advances, vol. 28, no. 6, pp. 770–781, 2010.
[55]  P. A. Volberding and S. G. Deeks, “Antiretroviral therapy and management of HIV infection,” The Lancet, vol. 376, no. 9734, pp. 49–62, 2010.
[56]  D. M. Moore, A. Awor, R. Downing et al., “CD4+ T-cell count monitoring does not accurately identify HIV-infected adults with virologie failure receiving antiretroviral therapy,” Journal of Acquired Immune Deficiency Syndromes, vol. 49, no. 5, pp. 477–484, 2008.
[57]  B. M. Branson, “State of the art for diagnosis of HIV infection,” Clinical Infectious Diseases, vol. 45, supplement 4, pp. S221–S225, 2007.
[58]  D. A. Anderson, S. M. Crowe, and M. Garcia, “Point-of-care testing,” Current HIV/AIDS Reports, vol. 8, no. 1, pp. 31–37, 2011.
[59]  S. Buttò, B. Suligoi, E. Fanales-Belasio, and M. Raimondo, “Laboratory diagnostics for HIV infection,” Annali dell'Istituto Superiore di Sanità, vol. 46, no. 1, pp. 24–33, 2010.
[60]  M. Vincent, Y. Xu, and H. Kong, “Helicase-dependent isothermal DNA amplification,” European Molecular Biology Organization Reports, vol. 5, no. 8, pp. 795–800, 2004.
[61]  P. M. Lizardi, X. Huang, Z. Zhu, P. Bray-Ward, D. C. Thomas, and D. C. Ward, “Mutation detection and single-molecule counting using isothermal rolling-circle amplification,” Nature Genetics, vol. 19, no. 3, pp. 225–232, 1998.
[62]  J. Van Ness, L. K. Van Ness, and D. J. Galas, “Isothermal reactions for the amplification of oligonucleotides,” Proceedings of the National Academy of Sciences of the United States of America, vol. 100, no. 8, pp. 4504–4509, 2003.
[63]  K. A. Curtis, D. L. Rudolph, and S. M. Owen, “Rapid detection of HIV-1 by reverse-transcription, loop-mediated isothermal amplification (RT-LAMP),” Journal of Virological Methods, vol. 151, no. 2, pp. 264–270, 2008.
[64]  K. A. Curtis, D. L. Rudolph, and S. M. Owen, “Sequence-specific detection method for reverse transcription, loop-mediated isothermal amplification of HIV-1,” Journal of Medical Virology, vol. 81, no. 6, pp. 966–972, 2009.
[65]  K. A. Curtis, D. L. Rudolph, I. Nejad et al., “Isothermal amplification using a chemical heating device for point-of-care detection of HIV-1,” PLoS One, vol. 7, no. 2, Article ID e31432, 2012.
[66]  Y. Mori, T. Hirano, and T. Notomi, “Sequence specific visual detection of LAMP reactions by addition of cationic polymers,” BMC Biotechnology, vol. 6, article 3, 2006.
[67]  P. Francois, M. Tangomo, J. Hibbs et al., “Robustness of a loop-mediated isothermal amplification reaction for diagnostic applications,” FEMS Immunology and Medical Microbiology, vol. 62, no. 1, pp. 41–48, 2011.
[68]  C. Liu, E. Geva, M. Mauk et al., “An isothermal amplification reactor with an integrated isolation membrane for point-of-care detection of infectious diseases,” Analyst, vol. 136, no. 10, pp. 2069–2076, 2011.
[69]  M. Goto, E. Honda, A. Ogura, A. Nomoto, and K.-I. Hanaki, “Colorimetric detection of loop-mediated isothermal amplification reaction by using hydroxy naphthol blue,” BioTechniques, vol. 46, no. 3, pp. 167–172, 2009.
[70]  B. Deiman, P. Van Aarle, and P. Sillekens, “Characteristics and applications of nucleic acid sequence-based amplification (NASBA),” Applied Biochemistry and Biotechnology Part B, vol. 20, no. 2, pp. 163–179, 2002.
[71]  J. Jean, D. H. D'Souza, and L.-A. Jaykus, “Multiplex nucleic acid sequence-based amplification for simultaneous detection of several enteric viruses in model ready-to-eat foods,” Applied and Environmental Microbiology, vol. 70, no. 11, pp. 6603–6610, 2004.
[72]  O. Piepenburg, C. H. Williams, D. L. Stemple, and N. A. Armes, “DNA detection using recombination proteins,” PLoS Biology, vol. 4, article e204, no. 7, 2006.
[73]  T. Notomi, H. Okayama, H. Masubuchi et al., “Loop-mediated isothermal amplification of DNA,” Nucleic Acids Research, vol. 28, article E63, no. 12, 2000.
[74]  Y. Kimura, M. J. L. De Hoon, S. Aoki et al., “Optimization of turn-back primers in isothermal amplification,” Nucleic Acids Research, vol. 39, article e59, no. 9, 2011.
[75]  K. Nagamine, T. Hase, and T. Notomi, “Accelerated reaction by loop-mediated isothermal amplification using loop primers,” Molecular and Cellular Probes, vol. 16, no. 3, pp. 223–229, 2002.
[76]  Y. Mori, K. Nagamine, N. Tomita, and T. Notomi, “Detection of loop-mediated isothermal amplification reaction by turbidity derived from magnesium pyrophosphate formation,” Biochemical and Biophysical Research Communications, vol. 289, no. 1, pp. 150–154, 2001.
[77]  L. L. M. Poon, B. W. Y. Wong, E. H. T. Ma et al., “Sensitive and inexpensive molecular test for falciparum malaria: defecting Plasmodium falciparum DNA directly from heat-treated blood by loop-mediated isothermal amplification,” Clinical Chemistry, vol. 52, no. 2, pp. 303–306, 2006.
[78]  Y. Mori, M. Kitao, N. Tomita, and T. Notomi, “Real-time turbidimetry of LAMP reaction for quantifying template DNA,” Journal of Biochemical and Biophysical Methods, vol. 59, no. 2, pp. 145–157, 2004.
[79]  K. Nagamine, K. Watanabe, K. Ohtsuka, T. Hase, and T. Notomi, “Loop-mediated isothermal amplification reaction using a nondenatured template,” Clinical Chemistry, vol. 47, no. 9, pp. 1742–1743, 2001.
[80]  R. Suzuki, M. Ihira, Y. Enomoto et al., “Heat denaturation increases the sensitivity of the cytomegalovirus loop-mediated isothermal amplification method,” Microbiology and Immunology, vol. 54, no. 8, pp. 466–470, 2010.
[81]  A. Fire and S.-Q. Xu, “Rolling replication of short DNA circles,” Proceedings of the National Academy of Sciences of the United States of America, vol. 92, no. 10, pp. 4641–4645, 1995.
[82]  G. Kumar, E. Garnova, M. Reagin, and A. Vidali, “Improved multiple displacement amplification with φ29 DNA polymerase for genotyping of single human cells,” BioTechniques, vol. 44, no. 7, pp. 879–890, 2008.
[83]  J. Banér, M. Nilsson, M. Mendel-Hartvig, and U. Landegren, “Signal amplification of padlock probes by rolling circle replication,” Nucleic Acids Research, vol. 26, no. 22, pp. 5073–5078, 1998.
[84]  S. Henriksson, A.-L. Blomstr?m, L. Fuxler, C. Fossum, M. Berg, and M. Nilsson, “Development of an in situ assay for simultaneous detection of the genomic and replicative form of PCV2 using padlock probes and rolling circle amplification,” Virology Journal, vol. 8, article 37, 2011.
[85]  J. Melin, J. Jarvius, J. G?ransson, and M. Nilsson, “Homogeneous amplified single-molecule detection: characterization of key parameters,” Analytical Biochemistry, vol. 368, no. 2, pp. 230–238, 2007.
[86]  P. M. Williams, R. Li, N. A. Johnson, G. Wright, J.-D. Heath, and R. D. Gascoyne, “A novel method of amplification of FFPET-derived RNA enables accurate disease classification with microarrays,” Journal of Molecular Diagnostics, vol. 12, no. 5, pp. 680–686, 2010.
[87]  I. Biswas and P. Hsieh, “Identification and characterization of a thermostable MutS homolog from Thermus aquaticus,” Journal of Biological Chemistry, vol. 271, no. 9, pp. 5040–5048, 1996.
[88]  Y. Mitani, A. Lezhava, Y. Kawai et al., “Rapid SNP diagnostics using asymmetric isothermal amplification and a new mismatch-suppression technology,” Nature Methods, vol. 4, no. 3, pp. 257–262, 2007.
[89]  Y. Kimura, A. Oguchi-Katayama, Y. Kawai et al., “Tail variation of the folding primer affects the SmartAmp2 process differently,” Biochemical and Biophysical Research Communications, vol. 383, no. 4, pp. 455–459, 2009.
[90]  G. T. Walker, M. C. Little, J. G. Nadeau, and D. D. Shank, “Isothermal in vitro amplification of DNA by a restriction enzyme/DNA polymerase system,” Proceedings of the National Academy of Sciences of the United States of America, vol. 89, no. 1, pp. 392–396, 1992.
[91]  G. T. Walker, M. S. Fraiser, J. L. Schram, M. C. Little, J. G. Nadeau, and D. P. Malinowski, “Strand displacement amplification—an isothermal, in vitro DNA amplification technique,” Nucleic Acids Research, vol. 20, no. 7, pp. 1691–1696, 1992.
[92]  H. Jia, Z. Li, C. Liu, and Y. Cheng, “Ultrasensitive detection of microRNAs by exponential isothermal amplification,” Angewandte Chemie, vol. 49, no. 32, pp. 5498–5501, 2010.
[93]  M. Shimada, F. Hino, H. Sagawa, H. Mukai, K. Asada, and I. Kato, “Development of the detection system for Mycobacterium tuberculosis DNA by using the isothermal DNA amplification method ICAN,” Rinsho Byori, vol. 50, no. 5, pp. 528–532, 2002.
[94]  H. Mukai, T. Uemori, O. Takeda et al., “Highly efficient isothermal DNA amplification system using three elements of 5′-DNA-RNA-3′ chimeric primers, RNaseH and strand-displacing DNA polymerase,” Journal of Biochemistry, vol. 142, no. 2, pp. 273–281, 2007.
[95]  T. Uemori, H. Mukai, O. Takeda et al., “Investigation of the molecular mechanism of ICAN, a novel gene amplification method,” Journal of Biochemistry, vol. 142, no. 2, pp. 283–292, 2007.
[96]  L. Kulinsky, Z. Noroozi, and M. Madou, “Present technology and future trends in point-of-care microfluidic diagnostics,” Methods in Molecular Biology, vol. 949, pp. 3–23, 2013.
[97]  P. F. Mens, A. van Amerongen, P. Sawa, P. A. Kager, and H. D. F. H. Schallig, “Molecular diagnosis of malaria in the field: development of a novel 1-step nucleic acid lateral flow immunoassay for the detection of all 4 human Plasmodium spp. and its evaluation in Mbita, Kenya,” Diagnostic Microbiology and Infectious Disease, vol. 61, no. 4, pp. 421–427, 2008.
[98]  M. Bla?ková, M. Koets, P. Rauch, and A. van Amerongen, “Development of a nucleic acid lateral flow immunoassay for simultaneous detection of listeria spp. and listeria monocytogenes in food,” European Food Research and Technology, vol. 229, no. 6, pp. 867–874, 2009.
[99]  S. Assadollahi, C. Reininger, R. Palkovits, P. Pointl, and T. Schalkhammer, “From lateral flow devices to a novel nano-color microfluidic assay,” Sensors, vol. 9, no. 8, pp. 6084–6100, 2009.
[100]  W. R. Seitz, “Immunoassay labels based on chemiluminescence and bioluminescence,” Clinical Biochemistry, vol. 17, no. 2, pp. 120–125, 1984.
[101]  P. Corstjens, M. Zuiderwijk, A. Brink et al., “Use of up-converting phosphor reporters in lateral-flow assays to detect specific nucleic acid sequences: a rapid, sensitive DNA test to identify human papillomavirus type 16 infection,” Clinical Chemistry, vol. 47, no. 10, pp. 1885–1893, 2001.
[102]  X. Chu, X. Fu, K. Chen, G.-L. Shen, and R.-Q. Yu, “An electrochemical stripping metalloimmunoassay based on silver-enhanced gold nanoparticle label,” Biosensors and Bioelectronics, vol. 20, no. 9, pp. 1805–1812, 2005.
[103]  P. Von Lode, “Point-of-care immunotesting: approaching the analytical performance of central laboratory methods,” Clinical Biochemistry, vol. 38, no. 7, pp. 591–606, 2005.
[104]  A. Kupstat, M. U. Kumke, and N. Hildebrandt, “Toward sensitive, quantitative point-of-care testing (POCT) of protein markers: miniaturization of a homogeneous time-resolved fluoroimmunoassay for prostate-specific antigen detection,” Analyst, vol. 136, no. 5, pp. 1029–1035, 2011.
[105]  B. A. Rohrman, V. Leautaud, E. Molyneux, and R. R. Richards-Kortum, “A lateral flow assay for quantitative detection of amplified HIV-1 RNA,” PLoS One, vol. 7, Article ID e45611, 2012.
[106]  P. Craw and W. Balachandran, “Isothermal nucleic acid amplification technologies for point-of-care diagnostics: a critical review,” Lab on a Chip, vol. 12, no. 14, pp. 2469–2486, 2012.
[107]  F. Vitzthum and J. Bernhagen, “SYBR Green I: an ultrasensitive fluorescent dye for double-standed DNA quantification in solution and other applications,” Recent Research Developments in Analytical Biochemistry, vol. 2, pp. 65–93, 2002.
[108]  P. M. Holland, R. D. Abramson, R. Watson, and D. H. Gelfand, “Detection of specific polymerase chain reaction product by utilizing the 5′–3′ exonuclease activity of Thermus aquaticus DNA polymerase,” Proceedings of the National Academy of Sciences of the United States of America, vol. 88, no. 16, pp. 7276–7280, 1991.
[109]  A. W. Martinez, S. T. Phillips, G. M. Whitesides, and E. Carrilho, “Diagnostics for the developing world: microfluidic paper-based analytical devices,” Analytical Chemistry, vol. 82, no. 1, pp. 3–10, 2010.
[110]  W. Zhao, M. A. Brook, and Y. Li, “Design of gold nanoparticle-based colorimetric biosensing assays,” ChemBioChem, vol. 9, no. 15, pp. 2363–2371, 2008.
[111]  R. Koonakosit, “Semiquantitative determination of urinary glucose: comparison of home-made strip and routine tests,” Journal of the Medical Association of Thailand, vol. 83, no. 1, pp. S152–S160, 2000.
[112]  D. D. Liana, B. Raguse, J. J. Gooding, and E. Chow, “Recent advances in paper-based sensors,” Sensors, vol. 12, no. 9, pp. 11505–11526, 2012.
[113]  Y. Adiguzel and H. Kulah, “CMOS cell sensors for point-of-care diagnostics,” Sensors, vol. 12, no. 8, pp. 10042–10066, 2012.
[114]  K.-H. Lee, S.-H. Choi, J.-O. Lee, M.-J. Sohn, J.-B. Yoon, and G.-H. Cho, “An autonomous CMOS hysteretic sensor for the detection of desorption-free DNA hybridization,” Biosensors and Bioelectronics, vol. 26, no. 11, pp. 4591–4595, 2011.
[115]  J. M. Rothberg, W. Hinz, T. M. Rearick et al., “An integrated semiconductor device enabling non-optical genome sequencing,” Nature, vol. 475, no. 7356, pp. 348–352, 2011.
[116]  S. Wang, M. Esfahani, U. A. Gurkan, F. Inci, D. R. Kuritzkes, and U. Demirci, “Efficient on-chip isolation of HIV subtypes,” Lab on a Chip, vol. 12, no. 8, pp. 1508–1515, 2012.
[117]  S. Schumacher, J. Nestler, T. Otto et al., “Highly-integrated lab-on-chip system for point-of-care multiparameter analysis,” Lab on a Chip, vol. 12, no. 3, pp. 464–473, 2012.
[118]  V. Gubala, L. F. Harris, A. J. Ricco, M. X. Tan, and D. E. Williams, “Point of care diagnostics: status and future,” Analytical Chemistry, vol. 84, no. 2, pp. 487–515, 2012.
[119]  L. Gervais and E. Delamarche, “Toward one-step point-of-care immunodiagnostics using capillary-driven microfluidics and PDMS substrates,” Lab on a Chip, vol. 9, no. 23, pp. 3330–3337, 2009.
[120]  S. Fournier-Bidoz, T. L. Jennings, J. M. Klostranec et al., “Facile and rapid one-step mass preparation of quantum-dot barcodes,” Angewandte Chemie, vol. 47, no. 30, pp. 5577–5581, 2008.
[121]  T. Ruckstuhl, C. M. Winterflood, and S. Seeger, “Supercritical angle fluorescence immunoassay platform,” Analytical Chemistry, vol. 83, no. 6, pp. 2345–2350, 2011.
[122]  S. Tang and I. Hewlett, “Nanoparticle-based immunoassays for sensitive and early detection of HIV-1 capsid (p24) antigen,” The Journal of Infectious Diseases, vol. 201, supplement 1, pp. S59–S64, 2010.
[123]  T. Ohashi, K. Mawatari, K. Sato, M. Tokeshi, and T. Kitamori, “A micro-ELISA system for the rapid and sensitive measurement of total and specific immunoglobulin E and clinical application to allergy diagnosis,” Lab on a Chip, vol. 9, no. 7, pp. 991–995, 2009.
[124]  M. Hervás, M. A. López, and A. Escarpa, “Electrochemical immunosensing on board microfluidic chip platforms,” Trends in Analytical Chemistry, vol. 31, pp. 109–128, 2012.
[125]  D. W. Kimmel, G. Leblanc, M. E. Meschievitz, and D. E. Cliffel, “Electrochemical sensors and biosensors,” Analytical Chemistry, vol. 84, no. 2, pp. 685–707, 2012.
[126]  C. Li, L. A. Shutter, P.-M. Wu, C. H. Ahn, and R. K. Narayan, “Potential of a simple lab-on-a-tube for point-of-care measurements of multiple analytes,” Lab on a Chip, vol. 10, no. 11, pp. 1476–1479, 2010.
[127]  K. Teeparuksapun, M. Hedstr?m, E. Y. Wong, S. Tang, I. K. Hewlett, and B. Mattiasson, “Ultrasensitive detection of HIV-1 p24 antigen using nanofunctionalized surfaces in a capacitive immunosensor,” Analytical Chemistry, vol. 82, no. 20, pp. 8406–8411, 2010.
[128]  K. N. Han, C. A. Li, and G. H. Seong, “Microfluidic chips for immunoassays,” Analytical Chemistry, vol. 6, pp. 119–141, 2013.
[129]  R. McKendry, J. Zhang, Y. Arntz et al., “Multiple label-free biodetection and quantitative DNA-binding assays on a nanomechanical cantilever array,” Proceedings of the National Academy of Sciences of the United States of America, vol. 99, no. 15, pp. 9783–9788, 2002.
[130]  Y. Liu, X. Li, Z. Zhang, G. Zuo, Z. Cheng, and H. Yu, “Nanogram per milliliter-level immunologic detection of alpha-fetoprotein with integrated rotating-resonance microcantilevers for early-stage diagnosis of heptocellular carcinoma,” Biomedical Microdevices, vol. 11, no. 1, pp. 183–191, 2009.
[131]  A. L. Washburn, M. S. Luchansky, A. L. Bowman, and R. C. Bailey, “Quantitative, label-free detection of five protein biomarkers using multiplexed arrays of silicon photonic microring resonators,” Analytical Chemistry, vol. 82, no. 1, pp. 69–72, 2010.
[132]  S. X. Wang and A. M. Taratorin, Magnetic Information Storage Technology, Electromagnetism, Academic Press, 1st edition, 1999.
[133]  G. J. Zhang and Y. Ning, “Silicon nanowire biosensor and its applications in disease diagnostics: a review,” Analytica Chimica Acta, vol. 749, pp. 1–15, 2012.
[134]  X. Guo, “Surface plasmon resonance based biosensor technique: a review,” Journal of Biophotonics, vol. 5, no. 7, pp. 483–501, 2012.
[135]  P. S. Waggoner and H. G. Craighead, “Micro—and nanomechanical sensors for environmental, chemical, and biological detection,” Lab on a Chip, vol. 7, no. 10, pp. 1238–1255, 2007.
[136]  M.-I. Rocha-Gaso, C. March-Iborra, á. Montoya-Baides, and A. Arnau-Vives, “Surface generated acoustic wave biosensors for the detection of pathogens: a review,” Sensors, vol. 9, no. 7, pp. 5740–5769, 2009.
[137]  E. R. Hirst, Y. J. Yuan, W. L. Xu, and J. E. Bronlund, “Bond-rupture immunosensors—a review,” Biosensors and Bioelectronics, vol. 23, no. 12, pp. 1759–1768, 2008.
[138]  O. Mudanyali, S. Dimitrov, U. Sikora, S. Padmanabhan, I. Navruz, and A. Ozcan, “Integrated rapid-diagnostic-test reader platform on a cellphone,” Lab on a Chip, vol. 12, no. 15, pp. 2678–2686, 2012.
[139]  R. de la Rica and M. M. Stevens, “Plasmonic ELISA for the ultrasensitive detection of disease biomarkers with the naked eye,” Nature Nanotechnology, vol. 7, no. 12, pp. 821–824, 2012.
[140]  M. Schito, T. F. Peter, S. Cavanaugh et al., “Opportunities and challenges for cost-efficient implementation of new point-of-care diagnostics for HIV and tuberculosis,” Journal of Infectious Diseases, vol. 205, supplement 2, pp. S169–S180, 2012.
[141]  T. Crucitti, D. Taylor, G. Beelaert, K. Fransen, and L. Van Damme, “Performance of a rapid and simple HIV testing algorithm in a multicenter phase III microbicide clinical trial,” Clinical and Vaccine Immunology, vol. 18, no. 9, pp. 1480–1485, 2011.
[142]  S. J. Reynolds and J. Muwonga, “OraQuick Advance Rapid HIV-1/2 antibody test,” Expert Review of Molecular Diagnostics, vol. 4, no. 5, pp. 587–591, 2004.
[143]  K. A. Powers, C. Poole, A. E. Pettifor, and M. S. Cohen, “Rethinking the heterosexual infectivity of HIV-1: a systematic review and meta-analysis,” The Lancet Infectious Diseases, vol. 8, no. 9, pp. 553–563, 2008.
[144]  C. D. Pilcher, L. Eaton, S. Kalichman, C. Bisol, and R. Da Silva De Souza, “Approaching “HIV elimination”: interventions for acute HIV infection,” Current HIV/AIDS Reports, vol. 3, no. 4, pp. 160–168, 2006.
[145]  T. L. Creek, G. G. Sherman, J. Nkengasong et al., “Infant human immunodeficiency virus diagnosis in resource-limited settings: issues, technologies, and country experiences,” American Journal of Obstetrics and Gynecology, vol. 197, no. 3, pp. S64–S71, 2007.
[146]  B. H. Chohan, S. Emery, D. Wamalwa et al., “Evaluation of a single round polymerase chain reaction assay using dried blood spots for diagnosis of HIV-1 infection in infants in an African setting,” BMC Pediatrics, vol. 11, article 18, 2011.
[147]  E. Van Braeckel, M. Koutsoukos, P. Bourguignon, F. Clement, L. McNally, and G. Leroux-Roels, “Vaccine-induced HIV seropositivity: a problem on the rise,” Journal of Clinical Virology, vol. 50, no. 4, pp. 334–337, 2011.
[148]  M. L. Schito, M. P. D'Souza, S. M. Owen, and M. P. Busch, “Challenges for rapid molecular HIV diagnostics,” The Journal of Infectious Diseases, vol. 201, supplement 1, pp. S1–S6, 2010.
[149]  F. Rouet and C. Rouzioux, “The measurement of HIV-1 viral load in resource-limited settings: how and where?” Clinical Laboratory, vol. 53, no. 3-4, pp. 135–148, 2007.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413