22
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Multi-Dose Priming Regimens of PfSPZ Vaccine: Safety and Efficacy against Controlled Human Malaria Infection in Equatoguinean Adults

      research-article
      1 , 2 , 3 , 1 , 2 , 1 , 1 , 4 , 5 , 2 , 6 , 1 , 4 , 5 , 1 , 3 , 3 , 3 , 3 , 3 , 3 , 3 , 3 , 3 , 3 , 3 , 3 , 3 , 3 , 3 , 3 , 3 , 2 , 3 , 3 , 4 , 5 , 2 , 2 , 2 , 2 , 2 , 2 , 7 , 7 , 3 , 3 , 3 , 7 , 7 , 7 , 8 , 8 , 4 , 5 , 2 , 2 , 6 , 4 , 5 , 2 , 1 , 2 , * ,
      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.

          Plasmodium falciparum sporozoite (PfSPZ) Vaccine is composed of radiation-attenuated, aseptic, purified cryopreserved PfSPZ. Multiple clinical trials empirically assessing two to six doses have shown multi-dose priming (two to four doses the first week) to be optimal for protection in both 4- and 16-week regimens. In this randomized, double-blind, normal saline (NS) placebo-controlled trial, four groups (G) of 18- to 32-year-old Equatoguineans received multi-dose priming regimens with or without a delayed final dose at 4 or 16 weeks. The regimens were G1: days 1, 3, 5, 7, and 113; G2: days 1, 3, 5, and 7; G3: days 1, 3, 5, 7, and 29; and G4: days 1, 8, and 29. All doses were 9 × 10 5 PfSPZ. Tolerability, safety, immunogenicity, and vaccine efficacy (VE) against homologous controlled human malaria infection (CHMI) 6–7 weeks after vaccination were assessed to down-select the best regimen. All four regimens were safe and well tolerated, with no significant differences in adverse events (AEs) between vaccinees ( N = 84) and NS controls ( N = 20) or between regimens. Out of 19 controls, 13 developed Pf parasitemia by quantitative polymerase chain reaction (qPCR) after CHMI. Only the vaccine regimen administered on study days 1, 8, and 29 gave significant protection (7/21 vaccinees versus 13/19 controls infected, VE 51.3%, P = 0.03, Barnard’s test, two-tailed). There were no significant differences in antibodies against Pf circumsporozoite protein (PfCSP), a major SPZ antigen, between protected and nonprotected vaccinees or controls pre-CHMI. The six controls not developing Pf parasitemia had significantly higher antibodies to blood stage antigens Pf exported protein 1 (PfEXP1) and Pf merozoite surface protein 1 (PfMSP1) than the controls who developed parasitemia, suggesting naturally acquired immunity against Pf limited infections in controls. This study identified a safe, protective, 4-week, multi-dose prime vaccination regimen for assessment in future trials of PfSPZ Vaccine.

          Related collections

          Most cited references41

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

              Safety and efficacy of PfSPZ Vaccine against Plasmodium falciparum via direct venous inoculation in healthy malaria-exposed adults in Mali: a randomised, double-blind phase 1 trial.

              Plasmodium falciparum sporozite (PfSPZ) Vaccine is a metabolically active, non-replicating, whole malaria sporozoite vaccine that has been reported to be safe and protective against P falciparum controlled human malaria infection in malaria-naive individuals. We aimed to assess the safety and protective efficacy of PfSPZ Vaccine against naturally acquired P falciparum in malaria-experienced adults in Mali.
                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
                April 2022
                07 February 2022
                07 February 2022
                : 106
                : 4
                : 1215-1226
                Affiliations
                [ 1 ]Bagamoyo Research and Training Center, Ifakara Health Institute, Bagamoyo, Tanzania;
                [ 2 ]Sanaria Inc., Rockville, Maryland;
                [ 3 ]Ministry of Health and Social Welfare, Equatorial Guinea, Malabo, Equatorial Guinea;
                [ 4 ]Clinical Immunology Unit, Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland;
                [ 5 ]University of Basel, Basel, Switzerland;
                [ 6 ]Protein Potential LLC, Rockville, Maryland;
                [ 7 ]Medical Care Development International (MCDI), Silver Spring, Maryland;
                [ 8 ]Marathon EG Production, Ltd., Bioko Norte, Equatorial Guinea
                Author notes
                [* ]Address correspondence to Thomas L. Richie, Sanaria Inc., 9800 Medical Center Drive, Suite A209, Rockville, MD 20850. E-mail: trichie@ 123456sanaria.com

                Financial support: This work was supported by a public–private partnership, the Equatorial Guinea Malaria Vaccine Initiative EGMVI, 41 made up of the Government of Equatorial Guinea (EG) Ministry of Mines and Hydrocarbons, Marathon EG Production Limited, Noble Energy, Atlantic Methanol Production Company, and EG LNG.

                Disclosure: Sanaria Inc. manufactured PfSPZ Vaccine and PfSPZ Challenge and Protein Potential LLC is affiliated with Sanaria. Sanaria was the Sponsor of the clinical trial. L. W. P. C., N. K. C., E. R. J., Y. A., L. L., T. M., M. C. C., P. F. B., B. K. L. S, S. L. H., and T. L. R. are salaried, full-time employees of Sanaria Inc., the developer and sponsor of Sanaria PfSPZ Vaccine. V. P. and T. C. S. were salaried, full time employees of Sanaria Inc. at the time the trial was conducted. Thus, all authors associated with Sanaria or Protein Potential have potential conflicts of interest.

                Authors’ addresses: Said Abdallah Jongo, Bagamoyo Research and Training Center, Ifakara Health Institute, Bagamoyo, Tanzania, E-mail: sjongo@ 123456ihi.or.tz . L. W. Preston Church, Sanaria Inc., Rockville, MD, E-mail: lwpchurch@ 123456sanaria.com . Vicente Urbano Nsue Ndong Nchama, Ministry of Health and Social Welfare, Equatorial Guinea, Malabo, Equatorial Guinea, E-mail: viceurb2013@ 123456gmail.com . Ali Hamad, Bagamoyo Research and Training Center, Ifakara Health Institute, Bagamoyo, Tanzania, E-mail: ahamad@ 123456ihi.or.tz . Raul Chuquiyauri, Sanaria Inc., Rockville, MD, E-mail: rchuquiyauri@ 123456sanaria.com . Kamaka Ramadhani Kassim and Thabit Athuman, Bagamoyo Research and Training Center, Ifakara Health Institute, Bagamoyo, Tanzania, E-mails: kramadhani@ 123456ihi.or.tz and tathuman@ 123456ihi.or.tz . Anna Deal, Clinical Immunology Unit, Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland, and University of Basel, Basel, Switzerland, E-mail: annacadeal@ 123456gmail.com . Natasha KC, Sanaria Inc., Rockville, MD, and Protein Potential LLC, Rockville, MD, E-mail: nkc@ 123456sanaria.com . Ali Mtoro, Bagamoyo Research and Training Center, Ifakara Health Institute, Bagamoyo, Tanzania, E-mail: amtoro@ 123456ihi.or.tz . Maxmillian Mpina, Clinical Immunology Unit, Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland, and University of Basel, Basel, Switzerland, E-mail: mmpina@ 123456ihi.or.tz . Elizabeth Nyakarungu, Bagamoyo Research and Training Center, Ifakara Health Institute, Bagamoyo, Tanzania, E-mail: enyakarungu@ 123456ihi.or.tz . Gertrudis Owono Bidjimi, Marta Alene Owono, Escolástica Raquel Mansogo Mayé, Martín Eká Ondó Mangue, Genaro Nsué Nguema Okomo, Beltrán Ekuá Ntutumu Pasialo, Dolores Mbang Ondó Mandumbi, María-Silvia A. López Mikue, Fortunata Lobede Mochomuemue, Mariano Obiang Obono, Juan Carlos Momo Besahá, José Raso Bijeri, Gabriel Mbá Abegue, Yolanda Rimoy Veri, Ines Toichoa Bela, Federico Comsil Chochi, and José Enrique Lima Sánchez, Ministry of Health and Social Welfare, Equatorial Guinea, Malabo, Equatorial Guinea, E-mails: gertrudis.owono@ 123456gmail.com , owonoeyangmartaalene@ 123456gmail.com , mansogoe@ 123456gmail.com , mondo@ 123456mcd.org , genarimetro@ 123456gmail.com , bntutumu@ 123456mcd.org , doloresmbang1@ 123456yahoo.es , rusigr@ 123456live.com , flobede@ 123456mcd.org , mobiang@ 123456mcd.org , jmomo@ 123456mcd.org , jraso@ 123456mcd.org , avohona@ 123456gmail.com , yolandarimoy@ 123456gmail.com , itoichoa@ 123456mcd.org , fcomsil@ 123456mcd.org , and jlima@ 123456mcd.org . Vanessa Pencelli, Sanaria Inc., Rockville, MD, E-mail: vanessapencelli@ 123456gmail.com . Griselda Gayozo and José Antonio Esono Mbá Nlang, Ministry of Health and Social Welfare, Equatorial Guinea, Malabo, Equatorial Guinea, E-mails: grisel_gayozo@ 123456hotmail.com and jnlang@ 123456mcd.org . Tobias Schindler, Clinical Immunology Unit, Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland, and University of Basel, Basel, Switzerland, E-mail: tobias.schindler@ 123456swisstph.ch . Eric R. James, Yonas Abebe, Laurence Lemiale, Thomas C. Stabler, Tooba Murshedkar, and Mei-Chun Chen, Sanaria Inc., Rockville, MD, E-mails: ejames@ 123456sanaria.com , yabebe@ 123456sanaria.com , llemiale@ 123456sanaria.com , tstabler@ 123456sanaria.com , tmurshedkar@ 123456sanaria.com , and mchen@ 123456sanaria.com . Christopher Schwabe and Josea Ratsirarson, Medical Care Development International (MCDI), Silver Spring, MD, E-mails: cschwabe@ 123456mcd.org and jratsirarson@ 123456mcd.org . Matilde Riloha Rivas, Mitoha Ondo’o Ayekaba, and Diosdado Vicente Nsué Milang, Ministry of Health and Social Welfare, Equatorial Guinea, Malabo, Equatorial Guinea, E-mails: mrriloha@ 123456gmail.com , mitoha_ondo@ 123456yahoo.com , and diosdadonsuem@ 123456yahoo.es . Carlos Cortés Falla, Wonder P. Phiri, and Guillermo A. García, Medical Care Development International (MCDI), Silver Spring, MD, E-mails: ccortes@ 123456mcd.org , wphiri@ 123456mcd.org , and ggarcia@ 123456mcd.org . Carl D. Maas and Bonifacio Manguire Nlavo, Marathon EG Production, Ltd., Bioko Norte, Equatorial Guinea, E-mails: cdmaas@ 123456mailbox.sc.edu and manguirenl@ 123456marathonoil.com . Marcel Tanner, Clinical Immunology Unit, Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland, and University of Basel, Basel, Switzerland, E-mail: marcel.tanner@ 123456swisstph.ch . Peter F. Billingsley, Sanaria Inc., Rockville, MD, E-mail: pbillingsley@ 123456sanaria.com . B. Kim Lee Sim, Sanaria Inc., Rockville, MD, and Protein Potential LLC, Rockville, MD, E-mail: ksim@ 123456protpot.com . Claudia Daubenberger, Clinical Immunology Unit, Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland, and University of Basel, Basel, Switzerland, E-mail: claudia.daubenberger@ 123456swisstph.ch . Stephen L. Hoffman, Sanaria Inc., Rockville, MD, E-mail: slhoffman@ 123456sanaria.com . Salim Abdulla, Bagamoyo Research and Training Center, Ifakara Health Institute, Bagamoyo, Tanzania, E-mail: sabdulla@ 123456ihi.or.tz . Thomas L. Richie, Sanaria Inc., Rockville, MD, E-mail: trichie@ 123456sanaria.com .

                Article
                tpmd210942
                10.4269/ajtmh.21-0942
                8991366
                35130487
                1216edfb-60c7-4530-b378-bbf61cf7938e
                © 2022 by The American Society of Tropical Medicine and Hygiene

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

                History
                : 30 August 2021
                : 15 November 2021
                Page count
                Pages: 12
                Categories
                Research Article

                Infectious disease & Microbiology
                Infectious disease & Microbiology

                Comments

                Comment on this article

                scite_

                Similar content92

                Cited by13

                Most referenced authors927