Network


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

Hotspot


Dive into the research topics where Eric R. James is active.

Publication


Featured researches published by Eric R. James.


Science | 2013

Protection Against Malaria by Intravenous Immunization with a Nonreplicating Sporozoite Vaccine

Robert A. Seder; Lee Jah Chang; Mary E. Enama; Kathryn L. Zephir; Uzma N. Sarwar; Ingelise J. Gordon; LaSonji A. Holman; Eric R. James; Peter F. Billingsley; Anusha Gunasekera; Adam Richman; Sumana Chakravarty; Anita Manoj; Soundarapandian Velmurugan; Minglin Li; Adam Ruben; Tao Li; Abraham G. Eappen; Richard E. Stafford; Sarah Plummer; Cynthia S. Hendel; Laura Novik; Pamela Costner; Floreliz Mendoza; Jamie G. Saunders; Martha Nason; Jason H. Richardson; Jittawadee Murphy; Silas A. Davidson; Thomas L. Richie

Malaria Sporozoite Vaccine Each year, hundreds of millions of people are infected with Plasmodium falciparum, the mosquito-borne parasite that causes malaria. A preventative vaccine is greatly needed. Seder et al. (p. 1359, published online 8 August; see the Perspective by Good) now report the results from a phase I clinical trial where subjects were immunized intravenously with a whole, attenuated sporozoite vaccine. Three of 9 subjects who received four doses and zero of 6 subjects who received five doses of the vaccine went on to develop malaria after controlled malaria infection. Both antibody titers and cellular immune responses correlated positively with the dose of vaccine received, suggesting that both arms of the adaptive immune response may have participated in the observed protection. Intravenous immunization with an attenuated whole malaria sporozoite vaccine protected volunteers in a phase I clinical trial. [Also see Perspective by Good] Consistent, high-level, vaccine-induced protection against human malaria has only been achieved by inoculation of Plasmodium falciparum (Pf) sporozoites (SPZ) by mosquito bites. We report that the PfSPZ Vaccine—composed of attenuated, aseptic, purified, cryopreserved PfSPZ—was safe and wel-tolerated when administered four to six times intravenously (IV) to 40 adults. Zero of six subjects receiving five doses and three of nine subjects receiving four doses of 1.35 × 105 PfSPZ Vaccine and five of six nonvaccinated controls developed malaria after controlled human malaria infection (P = 0.015 in the five-dose group and P = 0.028 for overall, both versus controls). PfSPZ-specific antibody and T cell responses were dose-dependent. These data indicate that there is a dose-dependent immunological threshold for establishing high-level protection against malaria that can be achieved with IV administration of a vaccine that is safe and meets regulatory standards.


Science | 2011

Live Attenuated Malaria Vaccine Designed to Protect through Hepatic CD8+ T Cell Immunity

Judith E. Epstein; K. Tewari; Kirsten E. Lyke; B. K. L. Sim; Peter F. Billingsley; Matthew B. Laurens; Anusha Gunasekera; Sumana Chakravarty; Eric R. James; Martha Sedegah; Adam Richman; Soundarapandian Velmurugan; Sharina Reyes; Ming Lin Li; Kathryn Tucker; Adriana Ahumada; Adam Ruben; Tao Li; Richard E. Stafford; Abraham G. Eappen; C. Tamminga; Jason W. Bennett; Christian F. Ockenhouse; Jittawadee Murphy; J. Komisar; N. Thomas; Mark Loyevsky; Ashley Birkett; Christopher V. Plowe; C. Loucq

The efficacy of a sporozoite-based malaria vaccine is tested in humans, nonhuman primates, and mice. 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.


Human Vaccines | 2010

Development of a metabolically active, non-replicating sporozoite vaccine to prevent Plasmodium falciparum malaria

Stephen L. Hoffman; Peter F. Billingsley; Eric R. James; Adam Richman; Mark Loyevsky; Tao Li; Sumana Chakravarty; Anusha Gunasekera; Rana Chattopadhyay; Minglin Li; Richard E. Stafford; Adriana Ahumada; Judith E. Epstein; Martha Sedegah; Sharina Reyes; Thomas L. Richie; Kirsten E. Lyke; Robert Edelman; Matthew B. Laurens; Christopher V. Plowe; B. Kim Lee Sim

Immunization of volunteers by the bite of mosquitoes carrying radiation-attenuated Plasmodium falciparum sporozoites protects greater than 90% of such volunteers against malaria, if adequate numbers of immunizing biting sessions and sporozoite-infected mosquitoes are used. Nonetheless, until recently it was considered impossible to develop, license and commercialize a live, whole parasite P. falciparum sporozoite (PfSPZ) vaccine. In 2003 Sanaria scientists reappraised the potential impact of a metabolically active, non-replicating PfSPZ vaccine, and outlined the challenges to producing such a vaccine. Six years later, significant progress has been made in overcoming these challenges. This progress has enabled the manufacture and release of multiple clinical lots of a 1(st) generation metabolically active, non-replicating PfSPZ vaccine, the Sanaria PfSPZ Vaccine, submission of a successful Investigational New Drug application to the US Food and Drug Administration, and initiation of safety, immunogenicity and protective efficacy studies in volunteers in MD, US. Efforts are now focused on how best to achieve submission of a successful Biologics License Application and introduce the vaccine to the primary target population of African children in the shortest possible period of time. This will require implementation of a systematic, efficient clinical development plan. Short term challenges include optimizing the (1) efficiency and scale up of the manufacturing process and quality control assays, (2) dosage regimen and method of administration, (3) potency of the vaccine, and (4) logistics of delivering the vaccine to those who need it most, and finalizing the methods for vaccine stabilization and attenuation. A medium term goal is to design and build a facility for manufacturing highly potent and stable vaccine for pivotal Phase 3 studies and commercial launch.


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.


Nature Medicine | 2016

Protection against malaria at 1 year and immune correlates following PfSPZ vaccination

Andrew S. Ishizuka; Kirsten E. Lyke; Adam DeZure; Andrea A. Berry; Thomas L. Richie; Floreliz Mendoza; Mary E. Enama; Ingelise J. Gordon; Lee-Jah Chang; Uzma N Sarwar; Kathryn L. Zephir; LaSonji A. Holman; Eric R. James; Peter F. Billingsley; Anusha Gunasekera; Sumana Chakravarty; Anita Manoj; Minglin Li; Adam Ruben; Tao Li; Abraham G. Eappen; Richard E. Stafford; Natasha K C; Tooba Murshedkar; Hope DeCederfelt; Sarah Plummer; Cynthia S. Hendel; Laura Novik; Pamela Costner; Jamie G. Saunders

An attenuated Plasmodium falciparum (Pf) sporozoite (SPZ) vaccine, PfSPZ Vaccine, is highly protective against controlled human malaria infection (CHMI) 3 weeks after immunization, but the durability of protection is unknown. We assessed how vaccine dosage, regimen, and route of administration affected durable protection in malaria-naive adults. After four intravenous immunizations with 2.7 × 105 PfSPZ, 6/11 (55%) vaccinated subjects remained without parasitemia following CHMI 21 weeks after immunization. Five non-parasitemic subjects from this dosage group underwent repeat CHMI at 59 weeks, and none developed parasitemia. Although Pf-specific serum antibody levels correlated with protection up to 21–25 weeks after immunization, antibody levels waned substantially by 59 weeks. Pf-specific T cell responses also declined in blood by 59 weeks. To determine whether T cell responses in blood reflected responses in liver, we vaccinated nonhuman primates with PfSPZ Vaccine. Pf-specific interferon-γ-producing CD8 T cells were present at ∼100-fold higher frequencies in liver than in blood. Our findings suggest that PfSPZ Vaccine conferred durable protection to malaria through long-lived tissue-resident T cells and that administration of higher doses may further enhance protection.


Vaccine | 2015

Progress with Plasmodium falciparum sporozoite (PfSPZ)-based malaria vaccines.

Thomas L. Richie; Peter F. Billingsley; B. Kim Lee Sim; Eric R. James; Sumana Chakravarty; Judith E. Epstein; Kirsten E. Lyke; Benjamin Mordmüller; Pedro L. Alonso; Patrick E. Duffy; Ogobara K. Doumbo; Robert W. Sauerwein; Marcel Tanner; Salim Abdulla; Peter G. Kremsner; Robert A. Seder; Stephen L. Hoffman

Sanaria Inc. has developed methods to manufacture, purify and cryopreserve aseptic Plasmodium falciparum (Pf) sporozoites (SPZ), and is using this platform technology to develop an injectable PfSPZ-based vaccine that provides high-grade, durable protection against infection with Pf malaria. Several candidate vaccines are being developed and tested, including PfSPZ Vaccine, in which the PfSPZ are attenuated by irradiation, PfSPZ-CVac, in which fully infectious PfSPZ are attenuated in vivo by concomitant administration of an anti-malarial drug, and PfSPZ-GA1, in which the PfSPZ are attenuated by gene knockout. Forty-three research groups in 15 countries, organized as the International PfSPZ Consortium (I-PfSPZ-C), are collaborating to advance this program by providing intellectual, clinical, and financial support. Fourteen clinical trials of these products have been completed in the USA, Europe and Africa, two are underway and at least 12 more are planned for 2015–2016 in the US (four trials), Germany (2 trials), Tanzania, Kenya, Mali, Burkina Faso, Ghana and Equatorial Guinea. Sanaria anticipates application to license a first generation product as early as late 2017, initially to protect adults, and a year later to protect all persons >6 months of age for at least six months. Improved vaccine candidates will be advanced as needed until the following requirements have been met: long-term protection against natural transmission, excellent safety and tolerability, and operational feasibility for population-wide administration. Here we describe the three most developed whole PfSPZ vaccine candidates, associated clinical trials, initial plans for licensure and deployment, and long-term objectives for a final product suitable for mass administration to achieve regional malaria elimination and eventual global eradication.


American Journal of Tropical Medicine and Hygiene | 2014

Controlled Human Malaria Infection of Tanzanians by Intradermal Injection of Aseptic, Purified, Cryopreserved Plasmodium falciparum Sporozoites

Seif Shekalaghe; Mastidia Rutaihwa; Peter F. Billingsley; Mwajuma Chemba; Claudia Daubenberger; Eric R. James; Maximillian Mpina; Omar Juma; Tobias Schindler; Eric Huber; Anusha Gunasekera; Anita Manoj; Beatus Simon; Elizabeth Saverino; L. W. Preston Church; Cornelus C. Hermsen; Robert W. Sauerwein; Christopher V. Plowe; Meera Venkatesan; Philip Sasi; Omar Lweno; Paul Mutani; Ali Hamad; Ali Mohammed; Alwisa Urassa; Tutu Mzee; Debbie Padilla; Adam Ruben; B. Kim Lee Sim; Marcel Tanner

Controlled human malaria infection (CHMI) by mosquito bite has been used to assess anti-malaria interventions in > 1,500 volunteers since development of methods for infecting mosquitoes by feeding on Plasmodium falciparum (Pf) gametocyte cultures. Such CHMIs have never been used in Africa. Aseptic, purified, cryopreserved Pf sporozoites, PfSPZ Challenge, were used to infect Dutch volunteers by intradermal injection. We conducted a double-blind, placebo-controlled trial to assess safety and infectivity of PfSPZ Challenge in adult male Tanzanians. Volunteers were injected intradermally with 10,000 (N = 12) or 25,000 (N = 12) PfSPZ or normal saline (N = 6). PfSPZ Challenge was well tolerated and safe. Eleven of 12 and 10 of 11 subjects, who received 10,000 and 25,000 PfSPZ respectively, developed parasitemia. In 10,000 versus 25,000 PfSPZ groups geometric mean days from injection to Pf positivity by thick blood film was 15.4 versus 13.5 (P = 0.023). Alpha-thalassemia heterozygosity had no apparent effect on infectivity. PfSPZ Challenge was safe, well tolerated, and infectious.


PLOS ONE | 2013

Optimising Controlled Human Malaria Infection Studies Using Cryopreserved P. falciparum Parasites Administered by Needle and Syringe

Susanne H. Sheehy; Alexandra J. Spencer; Alexander D. Douglas; B. Kim Lee Sim; Rhea J. Longley; Nick J. Edwards; Ian D. Poulton; Domtila Kimani; Andrew R. Williams; Nicholas A. Anagnostou; Rachel Roberts; Simon Kerridge; Merryn Voysey; Eric R. James; Peter F. Billingsley; Anusha Gunasekera; Alison M. Lawrie; Stephen L. Hoffman; Adrian V. S. Hill

Background Controlled human malaria infection (CHMI) studies have become a routine tool to evaluate efficacy of candidate anti-malarial drugs and vaccines. To date, CHMI trials have mostly been conducted using the bite of infected mosquitoes, restricting the number of trial sites that can perform CHMI studies. Aseptic, cryopreserved P. falciparum sporozoites (PfSPZ Challenge) provide a potentially more accurate, reproducible and practical alternative, allowing a known number of sporozoites to be administered simply by injection. Methodology We sought to assess the infectivity of PfSPZ Challenge administered in different dosing regimens to malaria-naive healthy adults (nu200a=u200a18). Six participants received 2,500 sporozoites intradermally (ID), six received 2,500 sporozoites intramuscularly (IM) and six received 25,000 sporozoites IM. Findings Five out of six participants receiving 2,500 sporozoites ID, 3/6 participants receiving 2,500 sporozoites IM and 6/6 participants receiving 25,000 sporozoites IM were successfully infected. The median time to diagnosis was 13.2, 17.8 and 12.7 days for 2,500 sporozoites ID, 2,500 sporozoites IM and 25,000 sporozoites IM respectively (Kaplan Meier method; pu200a=u200a0.024 log rank test). Conclusions 2,500 sporozoites ID and 25,000 sporozoites IM have similar infectivities. Given the dose response in infectivity seen with IM administration, further work should evaluate increasing doses of PfSPZ Challenge IM to identify a dosing regimen that reliably infects 100% of participants. Trial Registration ClinicalTrials.gov NCT01465048


Frontiers in Microbiology | 2014

Evaluating controlled human malaria infection in Kenyan adults with varying degrees of prior exposure to Plasmodium falciparum using sporozoites administered by intramuscular injection.

Susanne H. Hodgson; Elizabeth Juma; Amina Salim; Charles Magiri; Domtila Kimani; Daniel Njenga; Alfred Muia; Andrew O Cole; Caroline Ogwang; Ken Awuondo; Brett Lowe; M Munene; Peter F. Billingsley; Eric R. James; Anusha Gunasekera; Bk Sim; Patricia Njuguna; Tommy Rampling; A Richman; Y Abebe; Gathoni Kamuyu; M Muthui; Sean C. Elias; Sassy Molyneux; Stephen Gerry; Alexander Macharia; Thomas N. Williams; Peter C. Bull; Adrian V. S. Hill; Faith Osier

Background: Controlled human malaria infection (CHMI) studies are a vital tool to accelerate vaccine and drug development. As CHMI trials are performed in a controlled environment, they allow unprecedented, detailed evaluation of parasite growth dynamics (PGD) and immunological responses. However, CHMI studies have not been routinely performed in malaria-endemic countries or used to investigate mechanisms of naturally-acquired immunity (NAI) to Plasmodium falciparum. Methods: We conducted an open-label, randomized CHMI pilot-study using aseptic, cryopreserved P. falciparum sporozoites (PfSPZ Challenge) to evaluate safety, infectivity and PGD in Kenyan adults with low to moderate prior exposure to P. falciparum (Pan African Clinical Trial Registry: PACTR20121100033272). Results: All participants developed blood-stage infection confirmed by quantitative polymerase chain reaction (qPCR). However one volunteer (110) remained asymptomatic and blood-film negative until day 21 post-injection of PfSPZ Challenge. This volunteer had a reduced parasite multiplication rate (PMR) (1.3) in comparison to the other 27 volunteers (median 11.1). A significant correlation was seen between PMR and screening anti-schizont Enzyme Linked Immunosorbent Assays (ELISA) OD (p = 0.044, R = −0.384) but not when volunteer 110 was excluded from the analysis (p = 0.112, R = −0.313). Conclusions: PfSPZ Challenge is safe and infectious in malaria-endemic populations and could be used to assess the efficacy of malaria vaccines and drugs in African populations. Whilst our findings are limited by sample size, our pilot study has demonstrated for the first time that NAI may impact on PMR post-CHMI in a detectable fashion, an important finding that should be evaluated in further CHMI studies.


JCI insight | 2017

Protection against Plasmodium falciparum malaria by PfSPZ Vaccine

Judith E. Epstein; Kristopher M. Paolino; Thomas L. Richie; Martha Sedegah; Alexandra Singer; Adam Ruben; Sumana Chakravarty; April Stafford; Richard C. Ruck; Abraham G. Eappen; Tao Li; Peter F. Billingsley; Anita Manoj; Joana C. Silva; Kara A. Moser; Robin Nielsen; Donna Tosh; Susan Cicatelli; Harini Ganeshan; Jessica Case; Debbie Padilla; Silas A. Davidson; Lindsey S Garver; Elizabeth Saverino; Tooba Murshedkar; Anusha Gunasekera; Patrick S. Twomey; Sharina Reyes; James E. Moon; Eric R. James

BACKGROUND: A radiation-attenuated Plasmodium falciparum (Pf) sporozoite (SPZ) malaria vaccine, PfSPZ Vaccine, protected 6 of 6 subjects (100%) against homologous Pf (same strain as in the vaccine) controlled human malaria infection (CHMI) 3 weeks after 5 doses administered intravenously. The next step was to assess protective efficacy against heterologous Pf (different from Pf in the vaccine), after fewer doses, and at 24 weeks. METHODS: The trial assessed tolerability, safety, immunogenicity, and protective efficacy of direct venous inoculation (DVI) of 3 or 5 doses of PfSPZ Vaccine in non-immune subjects. RESULTS: Three weeks after final immunization, 5 doses of 2.7 × 105 PfSPZ protected 12 of 13 recipients (92.3% [95% CI: 48.0, 99.8]) against homologous CHMI and 4 of 5 (80.0% [10.4, 99.5]) against heterologous CHMI; 3 doses of 4.5 × 105 PfSPZ protected 13 of 15 (86.7% [35.9, 98.3]) against homologous CHMI. Twenty-four weeks after final immunization, the 5-dose regimen protected 7 of 10 (70.0% [17.3, 93.3]) against homologous and 1 of 10 (10.0% [-35.8, 45.6]) against heterologous CHMI; the 3-dose regimen protected 8 of 14 (57.1% [21.5, 76.6]) against homologous CHMI. All 22 controls developed Pf parasitemia. PfSPZ Vaccine was well tolerated, safe, and easy to administer. No antibody or T cell responses correlated with protection. CONCLUSIONS: We have demonstrated for the first time to our knowledge that PfSPZ Vaccine can protect against a 3-week heterologous CHMI in a limited group of malaria-naive adult subjects. A 3-dose regimen protected against both 3-week and 24-week homologous CHMI (87% and 57%, respectively) in this population. These results provide a foundation for developing an optimized immunization regimen for preventing malaria. TRIAL REGISTRATION: ClinicalTrials.gov NCT02215707. FUNDING: Support was provided through the US Army Medical Research and Development Command, Military Infectious Diseases Research Program, and the Naval Medical Research Centers Advanced Medical Development Program.

Collaboration


Dive into the Eric R. James's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sumana Chakravarty

Indian Institute of Chemical Technology

View shared research outputs
Top Co-Authors

Avatar

Thomas L. Richie

Naval Medical Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

B. Kim Lee Sim

Naval Medical Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cynthia S. Hendel

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Floreliz Mendoza

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ingelise J. Gordon

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge