3
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Beyond Blood Smears: Qualification of Plasmodium 18S rRNA as a Biomarker for Controlled Human Malaria Infections

      research-article
      1 , 1 , 1 , 1 , 1 , 1 , 1 , 1 , 1 , 1 , 1 , 2 , 3 , 4 , 4 , 1 , 2 , 2 , 4 , 2 , 2 , 2 , 2 , 2 , 2 , 4 , 2 , 5 , 5 , 5 , 5 , 6 , 7 , 8 , 8 , 9 , 10 , 4 , 11 , 12 , 1 , 2 , 12 , 13 , *
      The American Journal of Tropical Medicine and Hygiene
      The American Society of Tropical Medicine and Hygiene

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract.

          18S rRNA is a biomarker that provides an alternative to thick blood smears in controlled human malaria infection (CHMI) trials. We reviewed data from CHMI trials at non-endemic sites that used blood smears and Plasmodium 18S rRNA/rDNA biomarker nucleic acid tests (NATs) for time to positivity. We validated a multiplex quantitative reverse transcription–polymerase chain reaction (qRT-PCR) for Plasmodium 18S rRNA, prospectively compared blood smears and qRT-PCR for three trials, and modeled treatment effects at different biomarker-defined parasite densities to assess the impact on infection detection, symptom reduction, and measured intervention efficacy. Literature review demonstrated accelerated NAT-based infection detection compared with blood smears (mean acceleration: 3.2–3.6 days). For prospectively tested trials, the validated Plasmodium 18S rRNA qRT-PCR positivity was earlier (7.6 days; 95% CI: 7.1–8.1 days) than blood smears (11.0 days; 95% CI: 10.3–11.8 days) and significantly preceded the onset of grade 2 malaria-related symptoms (12.2 days; 95% CI: 10.6–13.3 days). Discrepant analysis showed that the risk of a blood smear–positive, biomarker-negative result was negligible. Data modeling predicted that treatment triggered by specific biomarker-defined thresholds can differentiate complete, partial, and non-protective outcomes and eliminate many grade 2 and most grade 3 malaria-related symptoms post-CHMI. Plasmodium 18S rRNA is a sensitive and specific biomarker that can justifiably replace blood smears for infection detection in CHMI trials in non-endemic settings. This study led to biomarker qualification through the U.S. Food and Drug Administration for use in CHMI studies at non-endemic sites, which will facilitate biomarker use for the qualified context of use in drug and vaccine trials.

          Related collections

          Most cited references40

          • Record: found
          • Abstract: found
          • Article: not found

          Protection against a malaria challenge by sporozoite inoculation.

          An effective vaccine for malaria is urgently needed. Naturally acquired immunity to malaria develops slowly, and induction of protection in humans can be achieved artificially by the inoculation of radiation-attenuated sporozoites by means of more than 1000 infective mosquito bites. We exposed 15 healthy volunteers--with 10 assigned to a vaccine group and 5 assigned to a control group--to bites of mosquitoes once a month for 3 months while they were receiving a prophylactic regimen of chloroquine. The vaccine group was exposed to mosquitoes that were infected with Plasmodium falciparum, and the control group was exposed to mosquitoes that were not infected with the malaria parasite. One month after the discontinuation of chloroquine, protection was assessed by homologous challenge with five mosquitoes infected with P. falciparum. We assessed humoral and cellular responses before vaccination and before the challenge to investigate correlates of protection. All 10 subjects in the vaccine group were protected against a malaria challenge with the infected mosquitoes. In contrast, patent parasitemia (i.e., parasites found in the blood on microscopical examination) developed in all five control subjects. Adverse events were mainly reported by vaccinees after the first immunization and by control subjects after the challenge; no serious adverse events occurred. In this model, we identified the induction of parasite-specific pluripotent effector memory T cells producing interferon-gamma, tumor necrosis factor alpha, and interleukin-2 as a promising immunologic marker of protection. Protection against a homologous malaria challenge can be induced by the inoculation of intact sporozoites. (ClinicalTrials.gov number, NCT00442377.) 2009 Massachusetts Medical Society
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Live attenuated malaria vaccine designed to protect through hepatic CD8⁺ T cell immunity.

            Our goal is to develop a vaccine that sustainably prevents Plasmodium falciparum (Pf) malaria in ≥80% of recipients. Pf sporozoites (PfSPZ) administered by mosquito bites are the only immunogens shown to induce such protection in humans. Such protection is thought to be mediated by CD8(+) T cells in the liver that secrete interferon-γ (IFN-γ). We report that purified irradiated PfSPZ administered to 80 volunteers by needle inoculation in the skin was safe, but suboptimally immunogenic and protective. Animal studies demonstrated that intravenous immunization was critical for inducing a high frequency of PfSPZ-specific CD8(+), IFN-γ-producing T cells in the liver (nonhuman primates, mice) and conferring protection (mice). Our results suggest that intravenous administration of this vaccine will lead to the prevention of infection with Pf malaria.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Sterile protection against human malaria by chemoattenuated PfSPZ vaccine

              A highly protective malaria vaccine would greatly facilitate the prevention and elimination of malaria and containment of drug-resistant parasites. A high level (more than 90%) of protection against malaria in humans has previously been achieved only by immunization with radiation-attenuated Plasmodium falciparum (Pf) sporozoites (PfSPZ) inoculated by mosquitoes; by intravenous injection of aseptic, purified, radiation-attenuated, cryopreserved PfSPZ (‘PfSPZ Vaccine’); or by infectious PfSPZ inoculated by mosquitoes to volunteers taking chloroquine or mefloquine (chemoprophylaxis with sporozoites). We assessed immunization by direct venous inoculation of aseptic, purified, cryopreserved, non-irradiated PfSPZ (‘PfSPZ Challenge’) to malaria-naive, healthy adult volunteers taking chloroquine for antimalarial chemoprophylaxis (vaccine approach denoted as PfSPZ-CVac). Three doses of 5.12 × 104 PfSPZ of PfSPZ Challenge at 28-day intervals were well tolerated and safe, and prevented infection in 9 out of 9 (100%) volunteers who underwent controlled human malaria infection ten weeks after the last dose (group III). Protective efficacy was dependent on dose and regimen. Immunization with 3.2 × 103 (group I) or 1.28 × 104 (group II) PfSPZ protected 3 out of 9 (33%) or 6 out of 9 (67%) volunteers, respectively. Three doses of 5.12 × 104 PfSPZ at five-day intervals protected 5 out of 8 (63%) volunteers. The frequency of Pf-specific polyfunctional CD4 memory T cells was associated with protection. On a 7,455 peptide Pf proteome array, immune sera from at least 5 out of 9 group III vaccinees recognized each of 22 proteins. PfSPZ-CVac is a highly efficacious vaccine candidate; when we are able to optimize the immunization regimen (dose, interval between doses, and drug partner), this vaccine could be used for combination mass drug administration and a mass vaccination program approach to eliminate malaria from geographically defined areas.
                Bookmark

                Author and article information

                Journal
                Am J Trop Med Hyg
                Am. J. Trop. Med. Hyg
                tpmd
                tropmed
                The American Journal of Tropical Medicine and Hygiene
                The American Society of Tropical Medicine and Hygiene
                0002-9637
                1476-1645
                June 2019
                22 April 2019
                22 April 2019
                : 100
                : 6
                : 1466-1476
                Affiliations
                [1 ]Department of Laboratory Medicine, Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, Washington;
                [2 ]Center for Global Infectious Disease Research, Seattle Children’s Research Institute (formerly the Center for Infectious Disease Research), Seattle, Washington;
                [3 ]Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Washington, Seattle, Washington;
                [4 ]Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland;
                [5 ]Sanaria, Inc., Rockville, Maryland;
                [6 ]Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington;
                [7 ]Duke Global Health Institute, Duke University, Durham, North Carolina;
                [8 ]Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland;
                [9 ]Microbiology Service, Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland;
                [10 ]Partners in Diagnostics, Rockville, Maryland;
                [11 ]Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, Washington;
                [12 ]Seattle Malaria Clinical Trials Center, Fred Hutch Cancer Research Center, Seattle, Washington;
                [13 ]Department of Microbiology, University of Washington, Seattle, Washington
                Author notes
                [* ]Address correspondence to Sean C. Murphy, University of Washington, 750 Republican St., E633, Seattle, WA 98133. E-mail: murphysc@ 123456uw.edu

                Financial support: Funding was from the BMGF (OPP1133622 to S. M.), and the biomarker laboratory was supported in part by the NIH (AI-38858; P30 AI027757); the MC-001 study was supported by the Seattle Biomedical Research Institute; the MC-003 study was supported by the BMGF, which was involved in development and design of the study; and the NIH PfSPZ-CVac PYR study was supported by the National Institute of Allergy and Infectious Diseases and NIH (U01AI109700 to Sanaria).

                Ethics, Consent, and Permissions: The MC-001 and MC-003 studies were reviewed and approved by the Western IRB (WIRB), in collaboration with UW IRB. The NIH PfSPZ-CVac PYR study was reviewed and approved by the NIAID IRB. Volunteers were enrolled after obtaining informed consent using IRB-approved documents. The IRB-approved informed consent for all studies included the consent to publish.

                Availability of data and material: Additional Biomarker Qualification data reviews are available on the FDA website. 51 All sequence data were from publicly available sources through plasmodb.org.

                Authors’ addresses: Annette M. Seilie, Ming Chang, Brad C. Stone, Glenda Daza, Jose Ortega, Kurtis R. Cruz, Nahum Smith, Carolyn K. Wallis, and Sean C. Murphy, Department of Laboratory Medicine, Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, E-mails: amseilie@ 123456uw.edu , minchang@ 123456uw.edu , stoneb4@ 123456uw.edu , gdaza@ 123456uw.edu , jortega@ 123456uw.edu , kurtisc@ 123456uw.edu , nahums@ 123456uw.edu , ckw@ 123456uw.edu , and murphysc@ 123456uw.edu . Amelia E. Hanron, Yale School of Public Health, New Haven, CT, E-mail: ameliahanron@ 123456gmail.com . Zachary P. Billman, MD/PhD Program, School of Medicine, University of North Carolina, Chapel Hill, NC, E-mail: z.p.billman@ 123456gmail.com . Kevin Zhou, Seattle Children’s Research Institute, Seattle, WA, E-mail: kevinzhouemail@ 123456gmail.com . Tayla M. Olsen, Fred Hutchinson Cancer Research Center, Seattle, WA, E-mail: tolsen@ 123456fredhutch.org . Sara A. Healy, Jillian Neal, Sharon Wong-Madden, Jen C. C. Hume, and Patrick E. Duffy, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, E-mails: sara.healy@ 123456nih.gov , jillian.vanvliet@ 123456nih.gov , sharon.wong-madden@ 123456nih.gov , jennifer.hume@ 123456nih.gov , and patrick.duffy@ 123456nih.gov . Lisa Shelton, Curevo, Inc., Seattle, WA, E-mail: lisarshelton@ 123456gmail.com . Sebastian A. Mikolajczak and Matt Fishbaugher, Novartis Institutes for BioMedical Research, Emeryville, CA, E-mails: sebastian.mikolajczak@ 123456novartis.com and mefishbaugher@ 123456gmail.com . Tracie (VonGoedert) Mankowski, Ashley M. Vaughan, Stefan H. I. Kappe, and Will Betz, Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, E-mails: tracie.mankowski@ 123456seattlechildrens.org , ashley.vaughan@ 123456seattlechildrens.org , stefan.kappe@ 123456seattlechildrens.org , and will.betz@ 123456seattlechildrens.org . Mark Kennedy, Kite, El Segundo, CA, E-mail: kennedy@ 123456kitepharma.com . Angela K. Talley, Spero Therapeutics, Cambridge, MA, E-mail: angelatalleymd@ 123456gmail.com . Stephen L. Hoffman, Sumana Chakravarty, B. Kim Lee Sim, Thomas L. Richie, Sanaria, Inc., Rockville, MD, E-mails: slhoffman@ 123456sanaria.com , schakravarty@ 123456sanaria.com , ksim@ 123456sanaria.com , and trichie@ 123456sanaria.com . Anna Wald, Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA, E-mail: annawald@ 123456uw.edu . Christopher V. Plowe, Duke Global Health Institute, Duke University, Durham, NC, E-mail: chris.plowe@ 123456duke.edu . Kirsten E. Lyke and Matthew Adams, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, E-mails: klyke@ 123456som.umaryland.edu and madams@ 123456som.umaryland.edu . Gary A. Fahle, Microbiology Service, Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, E-mail: gfahle@ 123456cc.nih.gov . Elliot P. Cowan, Partners in Diagnostics, Rockville, MD, E-mail: elliot.cowan@ 123456partnersindiagnostics.com . James G. Kublin, Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, E-mail: jkublinc@ 123456fredhutch.org .

                [†]

                These authors contributed equally to this work.

                Article
                tpmd190094
                10.4269/ajtmh.19-0094
                6553913
                31017084
                5143ddc3-6b50-4d23-abea-8bca4845dd4c
                © The American Society of Tropical Medicine and Hygiene

                This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

                History
                : 30 January 2019
                : 21 March 2019
                Page count
                Pages: 11
                Categories
                Articles

                Infectious disease & Microbiology
                Infectious disease & Microbiology

                Comments

                Comment on this article