Network


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

Hotspot


Dive into the research topics where Karina Teelen is active.

Publication


Featured researches published by Karina Teelen.


The New England Journal of Medicine | 2009

Protection against a Malaria Challenge by Sporozoite Inoculation

Meta Roestenberg; Matthew McCall; Joost Hopman; Jorien Wiersma; Adrian J. F. Luty; Geert Jan van Gemert; Marga van de Vegte-Bolmer; Ben C. L. van Schaijk; Karina Teelen; Theo Arens; Lopke Spaarman; Quirijn de Mast; Will Roeffen; Georges Snounou; Laurent Rénia; Andre van der Ven; Cornelus C. Hermsen; Robert W. Sauerwein

BACKGROUND 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. METHODS 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. RESULTS 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. CONCLUSIONS Protection against a homologous malaria challenge can be induced by the inoculation of intact sporozoites. (ClinicalTrials.gov number, NCT00442377.)


The Lancet | 2011

Long-term protection against malaria after experimental sporozoite inoculation: an open-label follow-up study

Meta Roestenberg; Anne C. Teirlinck; Matthew McCall; Karina Teelen; Krystelle Nganou Makamdop; Jorien Wiersma; Theo Arens; Pieter Beckers; Geert-Jan van Gemert; Marga van de Vegte-Bolmer; Andre van der Ven; Adrian J. F. Luty; Cornelus C. Hermsen; Robert W. Sauerwein

BACKGROUND We have shown that immunity to infection with Plasmodium falciparum can be induced experimentally in malaria-naive volunteers through immunisation by bites of infected mosquitoes while simultaneously preventing disease with chloroquine prophylaxis. This immunity was associated with parasite-specific production of interferon γ and interleukin 2 by pluripotent effector memory cells in vitro. We aim to explore the persistence of protection and immune responses in the same volunteers. METHODS In an open-label study at the Radboud University Nijmegen Medical Centre (Nijmegen, Netherlands), from November to December, 2009, we rechallenged previously immune volunteers (28 months after immunisation) with the bites of five mosquitoes infected with P falciparum. Newly recruited malaria-naive volunteers served as infection controls. Our primary outcome was the detection of blood-stage parasitaemia by microscopy. We assessed the kinetics of parasitaemia with real-time quantitative PCR (rtPCR) and recorded clinical signs and symptoms. In-vitro production of interferon γ and interleukin 2 by effector memory T cells was studied after stimulation with sporozoites and red blood cells infected with P falciparum. Differences in cellular immune responses between the study groups were assessed with the Mann-Whitney test. This study is registered with ClinicalTrials.gov, number NCT00757887. FINDINGS Four of six immune volunteers were microscopically negative after rechallenge. rtPCR-based detection of blood-stage parasites in these individuals was negative throughout follow-up. Patent parasitaemia was delayed in the remaining two immunised volunteers. In-vitro assays showed the long-term persistence of parasite-specific pluripotent effector memory T-cell responses in protected volunteers. The four protected volunteers reported several mild to moderate adverse events, of which the most commonly reported symptom was headache (one to three episodes per volunteer). The two patients with delayed patency had adverse events similar to those in the control group. INTERPRETATION Artificially induced immunity lasts longer than generally recorded after natural exposure; providing a new avenue of research into the mechanisms of malaria immunity. FUNDING Dioraphte Foundation.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Protection against malaria after immunization by chloroquine prophylaxis and sporozoites is mediated by preerythrocytic immunity

Else M. Bijker; Guido J. H. Bastiaens; Anne C. Teirlinck; Geert-Jan van Gemert; Wouter Graumans; Marga van de Vegte-Bolmer; Rianne Siebelink-Stoter; Theo Arens; Karina Teelen; Wiebke Nahrendorf; Edmond J. Remarque; Will Roeffen; Annemieke Jansens; Dunja Zimmerman; Martijn W. Vos; Ben C. L. van Schaijk; Jorien Wiersma; Andre van der Ven; Quirijn de Mast; Lisette van Lieshout; Jaco J. Verweij; Cornelus C. Hermsen; Anja Scholzen; Robert W. Sauerwein

Volunteers immunized under chloroquine chemoprophylaxis with Plasmodium falciparum sporozoites (CPS) develop complete, long-lasting protection against homologous sporozoite challenge. Chloroquine affects neither sporozoites nor liver-stages, but kills only asexual forms in erythrocytes once released from the liver into the circulation. Consequently, CPS immunization exposes the host to antigens from both preerythrocytic and blood stages, and induced immunity might target either of these stages. We therefore explored the life cycle stage specificity of CPS-induced protection. Twenty-five malaria-naïve volunteers were enrolled in a clinical trial, 15 of whom received CPS immunization. Five immunized subjects and five controls received a sporozoite challenge by mosquito bites, whereas nine immunized and five control subjects received an i.v. challenge with P. falciparum-infected erythrocytes. The latter approach completely bypasses preerythrocytic stages, enabling a direct comparison of protection against either life cycle stage. CPS-immunized subjects (13 of 14) developed anticircumsporozoite antibodies, whereas only one volunteer generated minimal titers against typical blood-stage antigens. IgG from CPS-immunized volunteers did not inhibit asexual blood-stage growth in vitro. All CPS-immunized subjects (5 of 5) were protected against sporozoite challenge. In contrast, nine of nine CPS-immunized subjects developed parasitemia after blood-stage challenge, with identical prepatent periods and blood-stage multiplication rates compared with controls. Intravenously challenged CPS-immunized subjects showed earlier fever and increased plasma concentrations of inflammatory markers D-dimer, IFN-γ, and monokine induced by IFN-γ than i.v. challenged controls. The complete lack of protection against blood-stage challenge indicates that CPS-induced protection is mediated by immunity against preerythrocytic stages. However, evidence is presented for immune recognition of P. falciparum-infected erythrocytes, suggesting memory responses unable to generate functional immunity.


American Journal of Tropical Medicine and Hygiene | 2013

Controlled Human Malaria Infections by Intradermal Injection of Cryopreserved Plasmodium falciparum Sporozoites

Meta Roestenberg; Else M. Bijker; B. Kim Lee Sim; Peter F. Billingsley; Eric R. James; Guido J. H. Bastiaens; Anne C. Teirlinck; Anja Scholzen; Karina Teelen; Theo Arens; Andre van der Ven; Anusha Gunasekera; Sumana Chakravarty; Soundarapandian Velmurugan; Cornelus C. Hermsen; Robert W. Sauerwein; Stephen L. Hoffman

Controlled human malaria infection with sporozoites is a standardized and powerful tool for evaluation of malaria vaccine and drug efficacy but so far only applied by exposure to bites of Plasmodium falciparum (Pf)-infected mosquitoes. We assessed in an open label Phase 1 trial, infection after intradermal injection of respectively 2,500, 10,000, or 25,000 aseptic, purified, vialed, cryopreserved Pf sporozoites (PfSPZ) in three groups (N = 6/group) of healthy Dutch volunteers. Infection was safe and parasitemia developed in 15 of 18 volunteers (84%), 5 of 6 volunteers in each group. There were no differences between groups in time until parasitemia by microscopy or quantitative polymerase chain reaction, parasite kinetics, clinical symptoms, or laboratory values. This is the first successful infection by needle and syringe with PfSPZ manufactured in compliance with regulatory standards. After further optimization, the use of such PfSPZ may facilitate and accelerate clinical development of novel malaria drugs and vaccines.


PLOS ONE | 2008

Safety and Immunogenicity of a Recombinant Plasmodium falciparum AMA1 Malaria Vaccine Adjuvanted with Alhydrogel™, Montanide ISA 720 or AS02

Meta Roestenberg; Ed Remarque; Erik de Jonge; Rob Hermsen; Hildur E. Blythman; Odile Leroy; Egeruan B. Imoukhuede; Søren Jepsen; Opokua Ofori-Anyinam; Bart W. Faber; Clemens H. M. Kocken; Miranda Arnold; Vanessa Walraven; Karina Teelen; Will Roeffen; Quirijn de Mast; W. Ripley Ballou; Joe Cohen; Marie-Claude Dubois; Stéphane Ascarateil; Andre van der Ven; Alan W. Thomas; Robert W. Sauerwein

Background Plasmodium falciparum Apical Membrane Antigen 1 (PfAMA1) is a candidate vaccine antigen expressed by merozoites and sporozoites. It plays a key role in red blood cell and hepatocyte invasion that can be blocked by antibodies. Methodology/Principal Findings We assessed the safety and immunogenicity of recombinant PfAMA1 in a dose-escalating, phase Ia trial. PfAMA1 FVO strain, produced in Pichia pastoris, was reconstituted at 10 µg and 50 µg doses with three different adjuvants, Alhydrogel™, Montanide ISA720 and AS02 Adjuvant System. Six randomised groups of healthy male volunteers, 8–10 volunteers each, were scheduled to receive three immunisations at 4-week intervals. Safety and immunogenicity data were collected over one year. Transient pain was the predominant injection site reaction (80–100%). Induration occurred in the Montanide 50 µg group, resulting in a sterile abscess in two volunteers. Systemic adverse events occurred mainly in the AS02 groups lasting for 1–2 days. Erythema was observed in 22% of Montanide and 59% of AS02 group volunteers. After the second dose, six volunteers in the AS02 group and one in the Montanide group who reported grade 3 erythema (>50 mm) were withdrawn as they met the stopping criteria. All adverse events resolved. There were no vaccine-related serious adverse events. Humoral responses were highest in the AS02 groups. Antibodies showed activity in an in vitro growth inhibition assay up to 80%. Upon stimulation with the vaccine, peripheral mononuclear cells from all groups proliferated and secreted IFNγ and IL-5 cytokines. Conclusions/Significance All formulations showed distinct reactogenicity profiles. All formulations with PfAMA1 were immunogenic and induced functional antibodies. Trial Registration Clinicaltrials.gov NCT00730782


Parasite Immunology | 1996

Association between anti‐Pfs48/45 reactivity and P. falciparum transmission‐blocking activity in sera from Cameroon

Will Roeffen; B. Mulder; Karina Teelen; M. Bolmer; Wijnand Eling; Geoffrey Targett; P. J. A. Beckers; Robert W. Sauerwein

Pfs48/45, a sexual stage parasite protein doublet of P. falciparum, is a target of antibodies which inhibit the development of the parasite in the mosquito. Twenty‐eight (54%) out of 52 sera of gametocyte carriers from Cameroon reduced infectivity in the mosquito membrane feeding bioassay to less than 20% of the controls. These 52 sera were analysed by competition ELISAs for the presence of antibodies capable of competing the binding of six monoclonal antibodies (MoAbs) directed against five different epitopes on Pfs48/45. The percentage of these 52 Cameroon sera that competed with one of the MoAbs ranged from 13% (epitope I) to 33% (epitope IIc). Comparison of activity in the transmission‐blocking assay ( ≥80%) and in the Pfs48/45 competition ELISA show a relative specificity of 100% (24 of 24) and a relative sensitivity of 75% (21 of 28). Non‐blocking sera showed no competition with any of the MoAbs. These MoAbs were further used to study the diversity of epitopes among isolates of P. falciparum using a two‐site ELISA. MoAbs against epitope I, III and V reacted with four different isolates whereas epitope II could be subdivided into three epitopes. None of the isolates reacted with MoAb 3G12 (epitope IV). Using these four different isolates, the competition ELISA titre varies from 1/20 to 1/80 and no significant differences are found between the isolates except for epitope II where only three out of 11 positives for epitope IIa were also positive for epitope IIc.


Journal of Biological Chemistry | 2007

Epitope analysis of the malaria surface antigen pfs48/45 identifies a subdomain that elicits transmission blocking antibodies.

Nikolay S. Outchkourov; Adriaan M. W. Vermunt; Josephine Jansen; Anita M. Kaan; Will Roeffen; Karina Teelen; Edwin Lasonder; Anneke Braks; Marga van de Vegte-Bolmer; Li Yan Qiu; Robert W. Sauerwein; Hendrik G. Stunnenberg

Pfs48/45, a member of a Plasmodium-specific protein family, displays conformation-dependent epitopes and is an important target for malaria transmission-blocking (TB) immunity. To design a recombinant Pfs48/45-based TB vaccine, we analyzed the conformational TB epitopes of Pfs48/45. The Pfs48/45 protein was found to consist of a C-terminal six-cysteine module recognized by anti-epitope I antibodies, a middle four-cysteine module recognized by anti-epitopes IIb and III, and an N-terminal module recognized by anti-epitope V antibodies. Refolding assays identified that a fragment of 10 cysteines (10C), comprising the middle four-cysteine and the C-terminal six-cysteine modules, possesses superior refolding capacity. The refolded and partially purified 10C conformer elicited antibodies in mice that targeted at least two of the TB epitopes (I and III). The induced antibodies could block the fertilization of Plasmodium falciparum gametes in vivo in a concentration-dependent manner. Our results provide important insight into the structural organization of the Pfs48/45 protein and experimental support for a Pfs48/45-based subunit vaccine.


Malaria Journal | 2011

A cluster-randomized trial of mass drug administration with a gametocytocidal drug combination to interrupt malaria transmission in a low endemic area in Tanzania

Seif Shekalaghe; Chris Drakeley; Sven van den Bosch; Roel ter Braak; Charles Mwanziva; Salimu Semvua; Alutu Masokoto; Frank W. Mosha; Karina Teelen; Rob Hermsen; Lucy C. Okell; Roly Gosling; Robert W. Sauerwein; Teun Bousema

BackgroundEffective mass drug administration (MDA) with anti-malarial drugs can clear the human infectious reservoir for malaria and thereby interrupt malaria transmission. The likelihood of success of MDA depends on the intensity and seasonality of malaria transmission, the efficacy of the intervention in rapidly clearing all malaria parasite stages and the degree to which symptomatic and asymptomatic parasite carriers participate in the intervention. The impact of MDA with the gametocytocidal drug combination sulphadoxine-pyrimethamine (SP) plus artesunate (AS) plus primaquine (PQ, single dose 0.75 mg/kg) on malaria transmission was determined in an area of very low and seasonal malaria transmission in northern Tanzania.MethodsIn a cluster-randomized trial in four villages in Lower Moshi, Tanzania, eight clusters (1,110 individuals; cluster size 47- 209) were randomized to observed treatment with SP+AS+PQ and eight clusters (2,347 individuals, cluster size 55- 737) to treatment with placebo over three days. Intervention and control clusters were 1km apart; households that were located between clusters were treated as buffer zones where all individuals received SP+AS+PQ but were not selected for the evaluation. Passive case detection was done for the entire cohort and active case detection in 149 children aged 1-10 year from the intervention arm and 143 from the control arm. Four cross-sectional surveys assessed parasite carriage by microscopy and molecular methods during a five-month follow-up period.ResultsThe coverage rate in the intervention arm was 93.0% (1,117/1,201). Parasite prevalence by molecular detection methods was 2.2-2.7% prior to the intervention and undetectable during follow-up in both the control and intervention clusters. None of the slides collected during cross-sectional surveys had microscopically detectable parasite densities. Three clinical malaria episodes occurred in the intervention (n = 1) and control clusters (n = 2).ConclusionsThis study illustrates the possibility to achieve high coverage with a three-day intervention but also the difficulty in defining suitable outcome measures to evaluate interventions in areas of very low malaria transmission intensity. The decline in transmission intensity prior to the intervention made it impossible to assess the impact of MDA in the chosen study setting.Trial RegistrationClinicalTrials.gov: NCT00509015


Parasite Immunology | 2006

Transmission-reducing immunity is inversely related to age in Plasmodium falciparum gametocyte carriers.

Chris Drakeley; J. T. Bousema; N. I. J. Akim; Karina Teelen; Will Roeffen; A.H.W. Lensen; M. Bolmer; Wijnand Eling; Robert W. Sauerwein

Immunity to the sexual stages of Plasmodium falciparum is induced during natural infections and can significantly reduce the transmission of parasites to mosquitoes (transmission reducing activity; TRA) but little is known about how these responses develop with increasing age/exposure to malaria. Routinely TRA is measured in the standard membrane feeding assay (SMFA). Sera were collected from a total of 199 gametocyte carriers (median age 4 years, quartiles 2 and 9 years) near Ifakara, Tanzania; 128 samples were tested in the SMFA and generated TRA data classified as a reduction of > 50% and > 90% of transmission. TRA of > 50% was highest in young children (aged 1–2) with a significant decline with age (χ2 trend = 5·79, P = 0·016) and in logistic regression was associated with prevalence of antibodies to both Pfs230 and Pfs48/45 (OR 4·03, P = 0·011 and OR 2·43 P = 0·059, respectively). A TRA of > 90% reduction in transmission was not age related but was associated with antibodies to Pfs48/45 (OR 2·36, P = 0·055). Our data confirm that antibodies are an important component of naturally induced TRA. However, whilst a similar but small proportion of individuals at all ages have TRA > 90%, the gradual deterioration of TRA > 50% with age suggests decreased antibody concentration or affinity. This may be due to decreased exposure to gametocytes, probably as a result of increased asexual and/or gametocyte specific immunity.


The Journal of Infectious Diseases | 2013

NF135.C10: a new Plasmodium falciparum clone for controlled human malaria infections

Anne C. Teirlinck; Meta Roestenberg; Marga van de Vegte-Bolmer; Anja Scholzen; Moniek J. L. Heinrichs; Rianne Siebelink-Stoter; Wouter Graumans; Geert-Jan van Gemert; Karina Teelen; Martijn W. Vos; Krystelle Nganou-Makamdop; Steffen Borrmann; Yolanda P. A. Rozier; Marianne A. A. Erkens; Adrian J. F. Luty; Cornelus C. Hermsen; B. Kim Lee Sim; Lisette van Lieshout; Stephen L. Hoffman; Leo G. Visser; Robert W. Sauerwein

We established a new field clone of Plasmodium falciparum for use in controlled human malaria infections and vaccine studies to complement the current small portfolio of P. falciparum strains, primarily based on NF54. The Cambodian clone NF135.C10 consistently produced gametocytes and generated substantial numbers of sporozoites in Anopheles mosquitoes and diverged from NF54 parasites by genetic markers. In a controlled human malaria infection trial, 3 of 5 volunteers challenged by mosquitoes infected with NF135.C10 and 4 of 5 challenged with NF54 developed parasitemia as detected with microscopy. The 2 strains induced similar clinical signs and symptoms as well as cellular immunological responses. Clinical Trials Registration NCT01002833.

Collaboration


Dive into the Karina Teelen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Meta Roestenberg

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Will Roeffen

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar

Wouter Graumans

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar

Andre van der Ven

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar

Anja Scholzen

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Else M. Bijker

Radboud University Nijmegen

View shared research outputs
Researchain Logo
Decentralizing Knowledge