Network


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

Hotspot


Dive into the research topics where Joel N. Maslow is active.

Publication


Featured researches published by Joel N. Maslow.


npj Vaccines | 2016

In vivo protection against ZIKV infection and pathogenesis through passive antibody transfer and active immunisation with a prMEnv DNA vaccine

Karuppiah Muthumani; Bryan D. Griffin; Sangya Agarwal; Sagar Kudchodkar; Emma L. Reuschel; Hyeree Choi; Kimberly A. Kraynyak; Elizabeth K Duperret; Amelia Keaton; Christopher W. Chung; Yinho K Kim; Stephanie A. Booth; Trina Racine; Jian Yan; Matthew P. Morrow; Jingjing Jiang; Brian Lee; Stephanie Ramos; Kate E. Broderick; Charles Reed; Amir S. Khan; Laurent Humeau; Kenneth E. Ugen; Young Kyoung Park; Joel N. Maslow; Niranjan Y. Sardesai; J. Joseph Kim; Gary P. Kobinger; David B. Weiner

Significant concerns have been raised owing to the rapid global spread of infection and disease caused by the mosquito-borne Zika virus (ZIKV). Recent studies suggest that ZIKV can also be transmitted sexually, further increasing the exposure risk for this virus. Associated with this spread is a dramatic increase in cases of microcephaly and additional congenital abnormalities in infants of ZIKV-infected mothers, as well as a rise in the occurrence of Guillain Barre’ syndrome in infected adults. Importantly, there are no licensed therapies or vaccines against ZIKV infection. In this study, we generate and evaluate the in vivo efficacy of a novel, synthetic, DNA vaccine targeting the pre-membrane+envelope proteins (prME) of ZIKV. Following initial in vitro development and evaluation studies of the plasmid construct, mice and non-human primates were immunised with this prME DNA-based immunogen through electroporation-mediated enhanced DNA delivery. Vaccinated animals were found to generate antigen-specific cellular and humoral immunity and neutralisation activity. In mice lacking receptors for interferon (IFN)-α/β (designated IFNAR−/−) immunisation with this DNA vaccine induced, following in vivo viral challenge, 100% protection against infection-associated weight loss or death in addition to preventing viral pathology in brain tissue. In addition, passive transfer of non-human primate anti-ZIKV immune serum protected IFNAR−/− mice against subsequent viral challenge. This study in NHP and in a pathogenic mouse model supports the importance of immune responses targeting prME in ZIKV infection and suggests that additional research on this vaccine approach may have relevance for ZIKV control and disease prevention in humans.


The New England Journal of Medicine | 2017

Safety and Immunogenicity of an Anti–Zika Virus DNA Vaccine — Preliminary Report

Pablo Tebas; Christine Roberts; Kar Muthumani; Emma L. Reuschel; Sagar Kudchodkar; Faraz I. Zaidi; Scott White; Amir S. Khan; Trina Racine; Hyeree Choi; Jean D. Boyer; Young K. Park; Sylvie Trottier; Celine Remigio; Diane Krieger; Susan E. Spruill; Mark L. Bagarazzi; Gary P. Kobinger; David B. Weiner; Joel N. Maslow

Background Although Zika virus (ZIKV) infection is typically self-limiting, other associated complications such as congenital birth defects and the Guillain-Barré syndrome are well described. There are no approved vaccines against ZIKV infection. Methods In this phase 1, open-label clinical trial, we evaluated the safety and immunogenicity of a synthetic, consensus DNA vaccine (GLS-5700) encoding the ZIKV premembrane and envelope proteins in two groups of 20 participants each. The participants received either 1 mg or 2 mg of vaccine intradermally, with each injection followed by electroporation (the use of a pulsed electric field to introduce the DNA sequence into cells) at baseline, 4 weeks, and 12 weeks. Results The median age of the participants was 38 years, and 60% were women; 78% were white, and 22% black; in addition, 30% were Hispanic. At the interim analysis at 14 weeks (i.e., after the third dose of vaccine), no serious adverse events were reported. Local reactions at the vaccination site (e.g., injection-site pain, redness, swelling, and itching) occurred in approximately 50% of the participants. After the third dose of vaccine, binding antibodies (as measured on enzyme-linked immunosorbent assay) were detected in all the participants, with geometric mean titers of 1642 and 2871 in recipients of 1 mg and 2 mg of vaccine, respectively. Neutralizing antibodies developed in 62% of the samples on Vero-cell assay. On neuronal-cell assay, there was 90% inhibition of ZIKV infection in 70% of the serum samples and 50% inhibition in 95% of the samples. The intraperitoneal injection of postvaccination serum protected 103 of 112 IFNAR knockout mice (bred with deletion of genes encoding interferon-α and interferon-β receptors) (92%) that were challenged with a lethal dose of ZIKV-PR209 strain; none of the mice receiving baseline serum survived the challenge. Survival was independent of the neutralization titer. Conclusions In this phase 1, open-label clinical trial, a DNA vaccine elicited anti-ZIKV immune responses. Further studies are needed to better evaluate the safety and efficacy of the vaccine. (Funded by GeneOne Life Science and others; ZIKA-001 ClinicalTrials.gov number, NCT02809443 .).


Nature Communications | 2017

DNA vaccination protects mice against Zika virus-induced damage to the testes

Bryan D. Griffin; Kar Muthumani; Bryce M. Warner; Anna Majer; Mable Hagan; Jonathan Audet; Derek R. Stein; Charlene Ranadheera; Trina Racine; Marc-Antoine de La Vega; Jocelyne Piret; Stephanie Kucas; Kaylie N. Tran; Kathy L. Frost; Christine De Graff; Geoff Soule; Leanne Scharikow; Jennifer Scott; Gordon McTavish; Valerie Smid; Young K. Park; Joel N. Maslow; Niranjan Y. Sardesai; J. Joseph Kim; Xiaojian Yao; Alexander Bello; Robbin Lindsay; Guy Boivin; Stephanie A. Booth; Darwyn Kobasa

Zika virus (ZIKV) is an emerging pathogen causally associated with serious sequelae in fetuses, inducing fetal microcephaly and other neurodevelopment defects. ZIKV is primarily transmitted by mosquitoes, but can persist in human semen and sperm, and sexual transmission has been documented. Moreover, exposure of type-I interferon knockout mice to ZIKV results in severe damage to the testes, epididymis and sperm. Candidate ZIKV vaccines have shown protective efficacy in preclinical studies carried out in animal models, and several vaccines have entered clinical trials. Here, we report that administration of a synthetic DNA vaccine encoding ZIKV pre-membrane and envelope (prME) completely protects mice against ZIKV-associated damage to the testes and sperm and prevents viral persistence in the testes following challenge with a contemporary strain of ZIKV. These data suggest that DNA vaccination merits further investigation as a potential means to reduce ZIKV persistence in the male reproductive tract.


Vaccine | 2017

Vaccine development for emerging virulent infectious diseases

Joel N. Maslow

Abstract The recent outbreak of Zaire Ebola virus in West Africa altered the classical paradigm of vaccine development and that for emerging infectious diseases (EIDs) in general. In this paper, the precepts of vaccine discovery and advancement through pre-clinical and clinical assessment are discussed in the context of the recent Ebola virus, Middle East Respiratory Syndrome coronavirus (MERS-CoV), and Zika virus outbreaks. Clinical trial design for diseases with high mortality rates and/or high morbidity in the face of a global perception of immediate need and the factors that drive design in the face of a changing epidemiology are presented. Vaccines for EIDs thus present a unique paradigm to standard development precepts.


Scientific Reports | 2017

Preclinical evaluation of multi antigenic HCV DNA vaccine for the prevention of Hepatitis C virus infection

Hyo Jin Lee; Moonsup Jeong; Jooyeon Oh; Youngran Cho; Xuefei Shen; John E. Stone; Jian Yan; Zachary Rothkopf; Amir S. Khan; Byung Mun Cho; Young Kyoung Park; David B. Weiner; Woo-Chan Son; Joel N. Maslow

Direct-acting antiviral treatment for hepatitis C virus (HCV) infection is costly and does not protect from re-infection. For human and chimpanzees, recovery from acute HCV infection correlates with host CD4+ and CD8+ T cell responses. DNA plasmids targeting the HCV non-structural antigens NS3, NS4, and NS5, were previously reported to induce robust and sustained T cell responses in mice and primates. These plasmids were combined with a plasmid encoding cytokine IL-28B, together named as VGX-6150. The dose-dependent T cell response and safety of VGX-6150 administered intramuscularly and followed by electroporation was assessed in mice. Immune responses plateaued at 20 μg/dose with IL-28B demonstrating significant immunoadjuvant activity. Mice administered VGX-6150 at 40, 400, and 800 μg given either as a single injection or as 14 injections given bi-weekly over 26 weeks showed no vaccine related changes in any clinical parameter compared to placebo recipients. There was no evidence of VGX-6150 accumulation at the injection site or in any organ 1 month following the 14th vaccination. Based on these studies, the approximate lethal dose (ALD) exceeds 800 μg/dose and the NOAEL was 800 μg/dose in mouse. In conclusion, VGX-6150 appears safe and a promising preventive vaccine candidate for HCV infection.


Vaccine | 2018

Assay Challenges for Emerging Infectious Diseases: The Zika Experience

Christine Roberts; Joel N. Maslow

From the perspective of vaccine development, it is imperative to accurately diagnose target infections in order to exclude subjects with prior exposure from evaluations of vaccine effectiveness, to track incident infection during the course of a clinical trial and to differentiate immune reactions due to natural infections from responses that are vaccine related. When vaccine development is accelerated to a rapid pace in response to emerging infectious disease threats, the challenges to develop such diagnostic tools is even greater. This was observed through the recent expansion of Zika virus infections into the Western Hemisphere in 2014–2017. When initial Zika vaccine clinical trials were being designed and launched in response to the outbreak, there were no standardized sets of viral and immunological assays, and no approved diagnostic tests for Zika virus infection. The diagnosis of Zika virus infection is still an area of active research and development on many fronts. Here we review emerging infectious disease vaccine clinical assay development and trial execution with a special focus on the state of Zika virus clinical assays and diagnostics.


The Journal of Infectious Diseases | 2018

Zika-Induced Male Infertility in Mice Is Potentially Reversible and Preventable by Deoxyribonucleic Acid Immunization

Marc-Antoine de La Vega; Jocelyne Piret; Bryan D. Griffin; Chantal Rhéaume; Marie-Christine Venable; Julie Carbonneau; Christian Couture; Raquel Das Neves Almeida; Roland R. Tremblay; Kelly Grace Magalhães; Young K. Park; Christine Roberts; Joel N. Maslow; Niranjan Y. Sardesai; J. Joseph Kim; Kar Muthumani; David B. Weiner; Gary P. Kobinger; Guy Boivin

Background Zika virus (ZIKV) infection has been associated with prolonged viral excretion in human semen and causes testicular atrophy and infertility in 10-week-old immunodeficient mice. Methods Male IFNAR-/- mice, knockout for type I interferon receptor, were immunized with GLS-5700, a deoxyribonucleic acid-based vaccine, before a subcutaneous ZIKV challenge with 6 × 105 plaque-forming units at 13 weeks of age. On day 28 postinfection, testes and epididymides were collected in some mice for histological and functional analyses, whereas others were mated with naive female wild-type C57BL/6J. Results Although all mice challenged with ZIKV developed viremia, most of them were asymptomatic, showed no weight loss, and survived infection. On day 28 postinfection, none of the unvaccinated, infected mice (9 of 9) exhibited abnormal spermatozoa counts or motility. However, 33% (3 of 9) and 36% (4 of 11) of mated males from this group were infertile, from 2 independent studies. Contrarily, males from the noninfected and the vaccinated, infected groups were all fertile. On days 75 and 207 postinfection, partial recovery of fertility was observed in 66% (2 of 3) of the previously infertile males. Conclusions This study reports the effects of ZIKV infection on male fertility in a sublethal, immunodeficient mouse model and the efficacy of GLS-5700 vaccination in preventing male infertility.


Open Forum Infectious Diseases | 2017

ZIKA-001: Safety and Immunogenicity of an Engineered DNA Vaccine Against ZIKA virus infection

Pablo Tebas; Christine Roberts; Kar Muthumani; Emma L. Reuschel; Scott White; Amir S. Khan; Trina Racine; Hyeree Choi; Faraz I. Zaidi; Jean D. Boyer; Sagar Kudchodkar; Young Kyoung Park; Sylvie Trottier; Celine Remigio; Diane Krieger; Gary P. Kobinger; David B. Weiner; Joel N. Maslow

Abstract Background While Zika virus (ZIKV) infection is typically self-limited, congenital birth defects and Guillain-Barré syndrome are well-described. There are no therapies or vaccines against ZIKV infection. Methods ZIKA-001 is a phase I, open label, clinical trial designed to evaluate the safety, side effect profile, and immunogenicity of a synthetic, DNA vaccine (GLS-5700) targeting the pre-membrane+envelope proteins (prME) of the virus. Two groups of 20 participants received GLS-5700 at one of two dose levels: 1 mg or 2 mg DNA/dose at 0, 4, and 12 weeks. Vaccine was administered as 0.1 or 0.2 ml (1 or 2 mg) intradermal (ID) injection followed by electroporation (EP) with the CELLECTRA®-3P device Results The median age of the 40 participants was 38 (IQR 30–54) years; 60% were female 30% Latino and 78% white. No SAEs have been reported to date. Local minor AEs were injection site pain, redness, swelling and itching that occurred in half of the participants. Systemic adverse events were rare and included headache, myalgias, upper respiratory infections, fatigue/malaise and nausea. Four weeks after the first dose 25% vs. 60% of the participants in the 1 mg and 2 mg dose seroconverted. By week 6, 2 weeks after the second dose, the response was 65 and 84% respectively and 2 weeks after the third dose all participants in both dosing groups developed antibodies. At the end of the vaccination period over 60% of vaccinated person neutralized Zika virus in a vero cell assay and greater than 80% on neuronal cell targets. The protective efficacy of the antibodies generated by the vaccine was evaluated in the lethal IFNAR−/− mouse model. After the intraperitoneal administration of 0.1 ml of either baseline, week 14 serum or PBS the animals were challenged with 106 PFUs of ZIKV PR209 isolate. Whereas animals administered PBS (control) or baseline serum succumbed after a median of 5 days, those pretreated with week 14 serum from study participants survived suggesting that the humoral response generated by the vaccine is protective in this model. Conclusion Our trial shows for the first time in humans the safety and immunogenicity of an engineered DNA encoding consensus viral protein against ZIKV. Future studies will evaluate the effectiveness of the vaccine. Disclosures C. C. Roberts, GeneOne: Member, Salary. S. White, GeneOne: Member, Salary. A. S. Khan, Inovio: Employee and Shareholder, Salary and Stock. J. Boyer, Inovio: Employee and Shareholder, Salary and Stock. Y. K. Park, GeneOne: Board Member, CEO and Employee, Salary and Stock. S. Trottier, Canadian Institutes of Health Research: Investigator, Research grant. C. Remigio, GeneOne: Employee and Shareholder, Salary and Stock. G. P. Kobinger, GeneOne: Grant Investigator and Scientific Advisor, Grant recipient and Research support. D. Weiner, GeneOne: Grant Investigator and Scientific Advisor, Grant recipient, Licensing agreement or royalty and Stock. J. Maslow, GeneOne: Employee, Salary and Stock.


Molecular Therapy | 2016

436. Developing a Synthetic DNA Vaccine for an Emerging Pathogen - Middle East Respiratory Syndrome

Emma L. Reuschel; Karrupiah Muthumani; Kim Kraynyak; Colleen Tingey; Sagar Kudchodkar; Amir S. Khan; Niranjan Y. Sardesai; J. Joseph Kim; Joel N. Maslow; Young K. Park; Gary P. Kobinger; Darryl Falzarano; Heinrich Feldman; David B. Weiner

Background: Middle East Respiratory Syndrome (MERS) was first reported in 2012 in Saudi Arabia when a patient died from severe respiratory disease caused by a novel betacorona virus, MERS-CoV. Through November 2015, there have been 1618 confirmed global cases of MERS-CoV infection and 579 deaths reported to the World Health Organization (WHO). Currently, no vaccine or specific treatment is available and patients are treated with supportive care based on their clinical condition. While most MERS cases occur in and around Saudi Arabia, the recent outbreak in Korea highlights the potential for this disease to spread beyond the immediate region. A vaccine is needed to prevent future disease caused by MERS-CoV.Methods: A synthetic DNA MERS vaccine was generated using a consensus sequence of the MERS spike protein. Mice, dromedary camels, and non-human primates (NHP) were immunized with MERS-vaccine by intramuscular injection followed by electroporation. Cellular immune responses were measured by flow cytometry and IFNγ ELISpot. Humoral immune responses were measured by ELISA and neutralizing antibody (nAb) assay. Following immunization, NHPs were challenged with infectious MERS-CoV (EMC/2012) and monitored for signs of infection by clinical scoring and examinations. Viral load was measured by qRT-PCR and tissue sections were stained with H&E.Results: Immunization of mice with MERS-vaccine induced strong humoral and cellular responses. Mice produced strong binding antibody (bAb) titers and nAb titers. A strong, polyfunctional, CD4 and CD8 T cell response was detected against multiple epitopes across the MERS spike protein. Immunization of dromedary camels induced the production of MERS spike specific antibodies and nAbs. Immunization of NHPs induced strong bAb titers and nAb titers and a strong CD4 and CD8 T cell response. NHPs immunized with multiple vaccination regimens were also protected from signs of disease upon challenge with infectious MERS-CoV and showed a greater than 3 log reduction in viral load after challenge compared to unvaccinated animals.Conclusions: A consensus DNA MERS-vaccine was able to generate both a strong T cell and neutralizing antibody response in multiple animal models, including camels, a natural host for MERS-CoV and a probable source of human infection. MERS-vaccine was also able to protect NHPs from an infectious MERS-CoV challenge. These results demonstrate the promise of this consensus DNA MERS-vaccine as a candidate for vaccine development.


Microbes and Infection | 2018

Rapid response to an emerging infectious disease - Lessons learned from development of a synthetic DNA vaccine targeting Zika virus

Sagar Kudchodkar; Hyeree Choi; Emma L. Reuschel; Rianne Esquivel; Jackie Jin-Ah Kwon; Moonsup Jeong; Joel N. Maslow; Charles Reed; Scott White; J. Joseph Kim; Gary P. Kobinger; Pablo Tebas; David B. Weiner; Kar Muthumani

Collaboration


Dive into the Joel N. Maslow's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amir S. Khan

Center for Cell and Gene Therapy

View shared research outputs
Top Co-Authors

Avatar

Emma L. Reuschel

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

J. Joseph Kim

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Kar Muthumani

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Sagar Kudchodkar

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge