Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Nurse Educ Today ; 130: 105943, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37598637

ABSTRACT

BACKGROUND: Effective transition to practice for new graduate nurses (NGNs) is of national and international concern. Development of NGNs expands well beyond higher education studies and relies heavily on support during their first year of employment. Little is known of the differential development of NGNs, namely the trajectory of developing capability. AIM: This study differentiates NGN development during their first year of employment through appraisal of universal domains of nursing practice relevant to the international community. METHODS: Using a quantitative evaluation design NGN performance was appraised at 1-, 3-, and 9-months from February 2020 to November 2021, using a 23-item appraisal tool and accompanying behavioural cues organised around four universal domains of practice: professional values and behaviours; comprehensive nursing practice; organisational capabilities; personal growth and development; and a fifth domain specific to Australia, that is, legislative requirements. Workplace performance was appraised by clinical supervisors and numerically rated according to intensity of coaching required to meet requisite standards of practice. RESULTS: The shift in rating scores of intensity of coaching required, over three time periods across four key universal domains, were statistically significant (p < .001). These findings which indicate the intensity of required coaching for maintenance of standards reduced over the time period suggest advancing NGN capability. The domain representing professional values consistently rated the highest. The domain denoting legislative requirements largely flattened after three months. CONCLUSION: These findings corroborate the significant development of NGN capability during the first nine months of employment, especially during the initial three months. Furthermore, they provide empirical evidence that NGNs are most adept at demonstrating professional values; a recognised capability developed through employment during pre-registration studies. Discriminant data is of value to inform both targeted development of individual NGNs and when collated, the education needs of cohorts.


Subject(s)
Education, Nursing, Graduate , Humans , Employment , Workplace , Australia , Cues
2.
Front Immunol ; 14: 1072810, 2023.
Article in English | MEDLINE | ID: mdl-36911698

ABSTRACT

Cancer immunotherapy has demonstrated great promise with several checkpoint inhibitors being approved as the first-line therapy for some types of cancer, and new engineered cytokines such as Neo2/15 now being evaluated in many studies. In this work, we designed antibody-cytokine chimera (ACC) scaffolding cytokine mimetics on a full-length tumor-specific antibody. We characterized the pharmacokinetic (PK) and pharmacodynamic (PD) properties of first-generation ACC TA99-Neo2/15, which synergized with DLnano-vaccines to suppress in vivo melanoma proliferation and induced significant systemic cytokine activation. A novel second-generation ACC TA99-HL2-KOA1, with retained IL-2Rß/γ binding and attenuated but preserved IL-2Rα binding, induced lower systemic cytokine activation with non-inferior protection in murine tumor studies. Transcriptomic analyses demonstrated an upregulation of Type I interferon responsive genes, particularly ISG15, in dendritic cells, macrophages and monocytes following TA99-HL2-KOA1 treatment. Characterization of additional ACCs in combination with cancer vaccines will likely be an important area of research for treating melanoma and other types of cancer.


Subject(s)
Melanoma , Nanoparticles , Vaccines, DNA , Mice , Animals , Cytokines , Antibodies , DNA
3.
Nurse Educ Today ; 111: 105296, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35131562

ABSTRACT

OBJECTIVE: This integrative review of the literature explores potential associations between paid employment during students' pre-registration study period and development of workplace capabilities. The capacity to demonstrate attainment of standards of practice upon registration as a nurse is essential for the delivery of safe, quality care. The increasing shift, internationally, to higher education, concerns have been raised about limited time in practice settings and consequently nurses' capability upon employment. Extensive research has been conducted into student clinical placement models and graduates transition programs, but employment during students' pre-registration study has received little consideration. DESIGN: An integrative approach of peer reviewed articles. DATA SOURCES: A systematic search of the literature published between 1996 and 2021 across five electronic data bases; including Cumulative Index to Nursing and Allied Health Literature, Scopus, Medline, American Psychological Association and Education Resource Information Centre was conducted. REVIEW METHODS: Data was analysed according to the Whittemore and Knafl (2005) framework to maintain a methodical and meticulous approach. RESULTS: Fourteen studies differentiated graduates employed during their studies. Employment contributed to developing capabilities across four domains, namely, personal growth and development, comprehensive nursing practice, organisational capability and professional values and behaviours upon employment. CONCLUSION: Employment during pre-registration studies is associated with developing workplace capabilities. Opportunities to develop the capability of graduates should focus on the possibility of 'learning' during employment rather than merely a recruitment strategy.


Subject(s)
Employment , Students, Nursing , Humans , Learning , Students , Workplace
4.
Nat Commun ; 13(1): 695, 2022 02 04.
Article in English | MEDLINE | ID: mdl-35121758

ABSTRACT

HIV Envelope (Env) is the main vaccine target for induction of neutralizing antibodies. Stabilizing Env into native-like trimer (NLT) conformations is required for recombinant protein immunogens to induce autologous neutralizing antibodies(nAbs) against difficult to neutralize HIV strains (tier-2) in rabbits and non-human primates. Immunizations of mice with NLTs have generally failed to induce tier-2 nAbs. Here, we show that DNA-encoded NLTs fold properly in vivo and induce autologous tier-2 nAbs in mice. DNA-encoded NLTs also uniquely induce both CD4 + and CD8 + T-cell responses as compared to corresponding protein immunizations. Murine neutralizing antibodies are identified with an advanced sequencing technology. The structure of an Env-Ab (C05) complex, as determined by cryo-EM, identifies a previously undescribed neutralizing Env C3/V5 epitope. Beyond potential functional immunity gains, DNA vaccines permit in vivo folding of structured antigens and provide significant cost and speed advantages for enabling rapid evaluation of new HIV vaccines.


Subject(s)
AIDS Vaccines/immunology , Antibodies, Neutralizing/immunology , HIV Antibodies/immunology , HIV Infections/immunology , HIV-1/immunology , Vaccines, DNA/immunology , env Gene Products, Human Immunodeficiency Virus/immunology , AIDS Vaccines/administration & dosage , Animals , Antibodies, Neutralizing/ultrastructure , Antigens, Viral/immunology , Cell Line, Tumor , Cryoelectron Microscopy , Enzyme-Linked Immunospot Assay , Epitopes/immunology , HEK293 Cells , HIV Antibodies/ultrastructure , HIV Infections/prevention & control , HIV Infections/virology , HIV-1/physiology , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Mice, Inbred BALB C , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Vaccination/methods , Vaccines, DNA/administration & dosage , env Gene Products, Human Immunodeficiency Virus/chemistry
5.
Vaccines (Basel) ; 8(4)2020 Dec 07.
Article in English | MEDLINE | ID: mdl-33297341

ABSTRACT

Background: Several techniques are under investigation to improve the immunogenicity of HIV-1 DNA vaccine candidates. DNA vaccines are advantageous due to their ease of design, expression of multiple antigens, and safety. METHODS: The HVTN 098 trial assessed the PENNVAX®-GP DNA vaccine (encoding HIV env, gag, pol) administered with or without plasmid IL-12 at 0-, 1-, 3-, and 6-month timepoints via intradermal (ID) or intramuscular (IM) electroporation (EP) in healthy, adult participants. We report on safety, tolerability, and acceptability. RESULTS: HVTN 098 enrolled 94 participants: 85 received PENNVAX®-GP and nine received placebo. Visual analog scale (VAS) pain scores immediately after each vaccination were lower in the ID/EP than in the IM/EP group (medians 4.1-4.6 vs. 6-6.5, p < 0.01). IM/EP participants reported greater pain and/or tenderness at the injection site. Most ID/EP participants had skin lesions such as scabs/eschars, scars, and pigmentation changes, which resolved within 6 months in 51% of participants (24/55). Eighty-two percent of IM/EP and 92% of ID/EP participant survey responses showed acceptable levels of discomfort. CONCLUSIONS: ID/EP and IM/EP are distinct experiences; however, HIV-1 DNA vaccination by either route was safe, tolerable and acceptable by most study participants.

6.
Cancer Immunol Res ; 8(11): 1354-1364, 2020 11.
Article in English | MEDLINE | ID: mdl-32913042

ABSTRACT

Cytolytic T cells (CTL) play a pivotal role in surveillance against tumors. Induction of CTL responses by vaccination may be challenging, as it requires direct transduction of target cells or special adjuvants to promote cross-presentation. Here, we observed induction of robust CTL responses through electroporation-facilitated, DNA-launched nanoparticle vaccination (DLnano-vaccines). Electroporation was observed to mediate transient tissue apoptosis and macrophage infiltration, which were deemed essential to the induction of CTLs by DLnano-vaccines through a systemic macrophage depletion study. Bolus delivery of protein nano-vaccines followed by electroporation, however, failed to induce CTLs, suggesting direct in vivo production of nano-vaccines may be required. Following these observations, new DLnano-vaccines scaffolding immunodominant melanoma Gp100 and Trp2 epitopes were designed and shown to induce more potent and consistent epitope-specific CTL responses than the corresponding DNA monomeric vaccines or CpG-adjuvanted peptide vaccines. DNA, but not recombinant protein, nano-vaccinations induced CTL responses to these epitopes and suppressed melanoma tumor growth in mouse models in a CD8+ T-cell-dependent fashion. Further studies to explore the use of DLnano-vaccines against other cancer targets and the biology with which they induce CTLs are important.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Nanoparticles/metabolism , Neoplasms/immunology , T-Lymphocytes/immunology , Vaccines, DNA/therapeutic use , Animals , Female , Humans , Mice , Vaccines, DNA/pharmacology
7.
JCI Insight ; 5(13)2020 07 09.
Article in English | MEDLINE | ID: mdl-32437332

ABSTRACT

BACKGROUNDHVTN 098, a randomized, double-blind, placebo-controlled trial, evaluated the safety, tolerability, and immunogenicity of PENNVAX-GP HIV DNA vaccine, administered with or without plasmid IL-12 (pIL-12), via intradermal (ID) or intramuscular (IM) electroporation (EP) in healthy, HIV-uninfected adults. The study tested whether PENNVAX-GP delivered via ID/EP at one-fifth the dose could elicit equivalent immune responses to delivery via IM/EP and whether inclusion of pIL-12 provided additional benefit.METHODSParticipants received DNA encoding HIV-1 env/gag/pol in 3 groups: 1.6 mg ID (ID no IL-12 group, n = 20), 1.6 mg ID + 0.4 mg pIL-12 (ID + IL-12 group, n = 30), 8 mg IM + 1 mg pIL-12 (IM + IL-12 group, n = 30), or placebo (n = 9) via EP at 0, 1, 3, and 6 months. Results of cellular and humoral immunogenicity assessments are reported.RESULTSFollowing vaccination, the frequency of responders (response rate) to any HIV protein based on CD4+ T cells expressing IFN-γ or IL-2 was 96% for both the ID + IL-12 and IM + IL-12 groups; CD8+ T cell response rates were 64% and 44%, respectively. For ID delivery, the inclusion of pIL-12 increased CD4+ T cell response rate from 56% to 96%. The frequency of responders was similar (≥90%) for IgG binding antibody to gp140 consensus Env across all groups, but the magnitude was higher in the ID + IL-12 group compared with the IM + IL-12 group.CONCLUSIONPENNVAX-GP DNA induced robust cellular and humoral immune responses, demonstrating that immunogenicity of DNA vaccines can be enhanced by EP route and inclusion of pIL-12. ID/EP was dose sparing, inducing equivalent, or in some aspects superior, immune responses compared with IM/EP.TRIAL REGISTRATIONClinicalTrials.gov NCT02431767.FUNDINGThis work was supported by National Institute of Allergy and Infectious Diseases (NIAID), U.S. Public Health Service grants, an HIV Vaccine Design and Development Team contract, Integrated Preclinical/Clinical AIDS Vaccine Development Program, and an NIH award.


Subject(s)
AIDS Vaccines/immunology , DNA/immunology , HIV Infections/immunology , Vaccines, DNA/immunology , Adult , CD8-Positive T-Lymphocytes/immunology , HIV Infections/prevention & control , HIV-1/immunology , Humans , Immunity, Humoral/immunology , Middle Aged , United States , Vaccination/methods , Vaccines, DNA/genetics , Young Adult
8.
Adv Sci (Weinh) ; 7(8): 1902802, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32328416

ABSTRACT

Nanotechnologies are considered to be of growing importance to the vaccine field. Through decoration of immunogens on multivalent nanoparticles, designed nanovaccines can elicit improved humoral immunity. However, significant practical and monetary challenges in large-scale production of nanovaccines have impeded their widespread clinical translation. Here, an alternative approach is illustrated integrating computational protein modeling and adaptive electroporation-mediated synthetic DNA delivery, thus enabling direct in vivo production of nanovaccines. DNA-launched nanoparticles are demonstrated displaying an HIV immunogen spontaneously self-assembled in vivo. DNA-launched nanovaccines induce stronger humoral responses than their monomeric counterparts in both mice and guinea pigs, and uniquely elicit CD8+ effector T-cell immunity as compared to recombinant protein nanovaccines. Improvements in vaccine responses recapitulate when DNA-launched nanovaccines with alternative scaffolds and decorated antigen are designed and evaluated. Finally, evaluation of functional immune responses induced by DLnanovaccines demonstrates that, in comparison to control mice or mice immunized with DNA-encoded hemagglutinin monomer, mice immunized with a DNA-launched hemagglutinin nanoparticle vaccine fully survive a lethal influenza challenge, and have substantially lower viral load, weight loss, and influenza-induced lung pathology. Additional study of these next-generation in vivo-produced nanovaccines may offer advantages for immunization against multiple disease targets.

9.
J Clin Invest ; 130(2): 827-837, 2020 02 03.
Article in English | MEDLINE | ID: mdl-31697648

ABSTRACT

Interventions to prevent HIV-1 infection and alternative tools in HIV cure therapy remain pressing goals. Recently, numerous broadly neutralizing HIV-1 monoclonal antibodies (bNAbs) have been developed that possess the characteristics necessary for potential prophylactic or therapeutic approaches. However, formulation complexities, especially for multiantibody deliveries, long infusion times, and production issues could limit the use of these bNAbs when deployed, globally affecting their potential application. Here, we describe an approach utilizing synthetic DNA-encoded monoclonal antibodies (dmAbs) for direct in vivo production of prespecified neutralizing activity. We designed 16 different bNAbs as dmAb cassettes and studied their activity in small and large animals. Sera from animals administered dmAbs neutralized multiple HIV-1 isolates with activity similar to that of their parental recombinant mAbs. Delivery of multiple dmAbs to a single animal led to increased neutralization breadth. Two dmAbs, PGDM1400 and PGT121, were advanced into nonhuman primates for study. High peak-circulating levels (between 6 and 34 µg/ml) of these dmAbs were measured, and the sera of all animals displayed broad neutralizing activity. The dmAb approach provides an important local delivery platform for the in vivo generation of HIV-1 bNAbs and for other infectious disease antibodies.


Subject(s)
Antibodies, Neutralizing/pharmacology , HIV Antibodies/pharmacology , HIV-1/immunology , Animals , Antibodies, Monoclonal, Murine-Derived/genetics , Antibodies, Monoclonal, Murine-Derived/immunology , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/immunology , Female , HEK293 Cells , HIV Antibodies/genetics , HIV Antibodies/immunology , Humans , Mice , Mice, Inbred BALB C
10.
Nat Commun ; 10(1): 3753, 2019 08 21.
Article in English | MEDLINE | ID: mdl-31434895

ABSTRACT

Semen is the vehicle for virion dissemination in the female reproductive tract (FRT) in male-to-female HIV transmission. Recent data suggests that higher frequency semen exposure is associated with activation of anti-HIV mechanisms in HIV negative sex workers. Here, we use a non-human primate (NHP) model to show that repeated vaginal exposure to semen significantly reduces subsequent infection by repeated low-dose vaginal SIVmac251 challenge. Repeated semen exposures result in lower CCR5 expression in circulating CD4+ T-cells, as well as higher expression of Mx1 (in correlation with IFNε expression) and FoxP3 in the cervicovaginal mucosa, and increased infiltration of CD4+ T-cells. Establishing in vivo evidence of competing effects of semen on transmission impacts our basic understanding of what factors may determine HIV infectivity in humans. Our results clearly indicate that repeated semen exposure can profoundly modulate the FRT microenvironment, paradoxically promoting host resistance against HIV acquisition.


Subject(s)
Cervix Uteri/immunology , Mucous Membrane/immunology , Semen/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/transmission , Simian Immunodeficiency Virus/immunology , Vagina/immunology , Animals , CD4-Positive T-Lymphocytes , Cervix Uteri/virology , Cytokines/metabolism , Disease Models, Animal , Female , Forkhead Transcription Factors/metabolism , HIV Infections/immunology , HIV Infections/transmission , Humans , Macaca mulatta , Mucous Membrane/metabolism , Myxovirus Resistance Proteins/metabolism , Receptors, CCR5/metabolism , Vagina/virology
11.
Lancet Infect Dis ; 19(9): 1013-1022, 2019 09.
Article in English | MEDLINE | ID: mdl-31351922

ABSTRACT

BACKGROUND: Middle East respiratory syndrome (MERS) coronavirus causes a highly fatal lower-respiratory tract infection. There are as yet no licensed MERS vaccines or therapeutics. This study (WRAIR-2274) assessed the safety, tolerability, and immunogenicity of the GLS-5300 MERS coronavirus DNA vaccine in healthy adults. METHODS: This study was a phase 1, open-label, single-arm, dose-escalation study of GLS-5300 done at the Walter Reed Army Institute for Research Clinical Trials Center (Silver Spring, MD, USA). We enrolled healthy adults aged 18-50 years; exclusion criteria included previous infection or treatment of MERS. Eligible participants were enrolled sequentially using a dose-escalation protocol to receive 0·67 mg, 2 mg, or 6 mg GLS-5300 administered by trained clinical site staff via a single intramuscular 1 mL injection at each vaccination at baseline, week 4, and week 12 followed immediately by co-localised intramuscular electroporation. Enrolment into the higher dose groups occurred after a safety monitoring committee reviewed the data following vaccination of the first five participants at the previous lower dose in each group. The primary outcome of the study was safety, assessed in all participants who received at least one study treatment and for whom post-dose study data were available, during the vaccination period with follow-up through to 48 weeks after dose 3. Safety was measured by the incidence of adverse events; administration site reactions and pain; and changes in safety laboratory parameters. The secondary outcome was immunogenicity. This trial is registered at ClinicalTrials.gov (number NCT02670187) and is completed. FINDINGS: Between Feb 17 and July 22, 2016, we enrolled 75 individuals and allocated 25 each to 0·67 mg, 2 mg, or 6 mg GLS-5300. No vaccine-associated serious adverse events were reported. The most common adverse events were injection-site reactions, reported in 70 participants (93%) of 75. Overall, 73 participants (97%) of 75 reported at least one solicited adverse event; the most common systemic symptoms were headache (five [20%] with 0·67 mg, 11 [44%] with 2 mg, and seven [28%] with 6 mg), and malaise or fatigue (five [20%] with 0·67 mg, seven [28%] with 2 mg, and two [8%] with 6 mg). The most common local solicited symptoms were administration site pain (23 [92%] with all three doses) and tenderness (21 [84%] with all three doses). Most solicited symptoms were reported as mild (19 [76%] with 0·67 mg, 20 [80%] with 2 mg, and 17 [68%] with 6 mg) and were self-limiting. Unsolicited symptoms were reported for 56 participants (75%) of 75 and were deemed treatment-related for 26 (35%). The most common unsolicited adverse events were infections, occurring in 27 participants (36%); six (8%) were deemed possibly related to study treatment. There were no laboratory abnormalities of grade 3 or higher that were related to study treatment; laboratory abnormalities were uncommon, except for 15 increases in creatine phosphokinase in 14 participants (three participants in the 0·67 mg group, three in the 2 mg group, and seven in the 6 mg group). Of these 15 increases, five (33%) were deemed possibly related to study treatment (one in the 2 mg group and four in the 6 mg group). Seroconversion measured by S1-ELISA occurred in 59 (86%) of 69 participants and 61 (94%) of 65 participants after two and three vaccinations, respectively. Neutralising antibodies were detected in 34 (50%) of 68 participants. T-cell responses were detected in 47 (71%) of 66 participants after two vaccinations and in 44 (76%) of 58 participants after three vaccinations. There were no differences in immune responses between dose groups after 6 weeks. At week 60, vaccine-induced humoral and cellular responses were detected in 51 (77%) of 66 participants and 42 (64%) of 66, respectively. INTERPRETATION: The GLS-5300 MERS coronavirus vaccine was well tolerated with no vaccine-associated serious adverse events. Immune responses were dose-independent, detected in more than 85% of participants after two vaccinations, and durable through 1 year of follow-up. The data support further development of the GLS-5300 vaccine, including additional studies to test the efficacy of GLS-5300 in a region endemic for MERS coronavirus. FUNDING: US Department of the Army and GeneOne Life Science.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , DNA, Viral/immunology , Middle East Respiratory Syndrome Coronavirus/immunology , Viral Vaccines/immunology , Adult , Fatigue/chemically induced , Female , Headache/chemically induced , Humans , Immunity, Cellular , Injection Site Reaction , Male , Viral Vaccines/administration & dosage , Viral Vaccines/adverse effects , Young Adult
12.
Infect Immun ; 87(8)2019 08.
Article in English | MEDLINE | ID: mdl-31182618

ABSTRACT

Vaccination remains one of the greatest medical breakthroughs in human history and has resulted in the near eradication of many formerly lethal diseases in many countries, including the complete eradication of smallpox. However, there remain a number of diseases for which there are no or only partially effective vaccines. There are numerous hurdles in vaccine development, of which knowing the appropriate immune response to target is one of them. Recently, tissue-resident T cells have been shown to mediate high levels of protection for several infections, although the best way to induce these cells is still unclear. Here we compare the ability to generate skin-resident T cells in sites distant from the immunization site following intramuscular and intradermal injection using optimized synthetic DNA vaccines. We found that mice immunized intradermally with a synthetic consensus DNA HIV envelope vaccine by electroporation (EP) are better able to maintain durable antigen-specific cellular responses in the skin than mice immunized by the intramuscular route. We extended these studies by delivering a synthetic DNA vaccine encoding Leishmania glycosomal phosphoenolpyruvate carboxykinase (PEPCK) by EP and again found that the intradermal route was superior to the intramuscular route for generating skin-resident PEPCK-specific T cells. We observed that when challenged with Leishmania major parasites, mice immunized intradermally exhibited significant protection, while mice immunized intramuscularly did not. The protection seen in intradermally vaccinated mice supports the viability of this platform not only to generate skin-resident T cells but also to promote durable protective immune responses at relevant tissue sites.


Subject(s)
Leishmania major/immunology , Leishmaniasis, Cutaneous/prevention & control , Protozoan Vaccines/immunology , Skin/immunology , T-Lymphocytes/immunology , Vaccination , Vaccines, DNA/immunology , Animals , Female , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL
13.
Vaccines (Basel) ; 7(2)2019 May 22.
Article in English | MEDLINE | ID: mdl-31121939

ABSTRACT

Identification of novel molecular adjuvants which can boost and enhance vaccine-mediated immunity and provide dose-sparing potential against complex infectious diseases and for immunotherapy in cancer is likely to play a critical role in the next generation of vaccines. Given the number of challenging targets for which no or only partial vaccine options exist, adjuvants that can address some of these concerns are in high demand. Here, we report that a designed truncated Interleukin-36 gamma (IL-36 gamma) encoded plasmid can act as a potent adjuvant for several DNA-encoded vaccine targets including human immunodeficiency virus (HIV), influenza, and Zika in immunization models. We further show that the truncated IL-36 gamma (opt-36γt) plasmid provides improved dose sparing as it boosts immunity to a suboptimal dose of a Zika DNA vaccine, resulting in potent protection against a lethal Zika challenge.

14.
JCI Insight ; 4(8)2019 04 18.
Article in English | MEDLINE | ID: mdl-30996140

ABSTRACT

Specific antibody therapy, including mAbs and bispecific T cell engagers (BiTEs), are important new tools for cancer immunotherapy. However, these approaches are slow to develop and may be limited in their production, thus restricting the patients who can access these treatments. BiTEs exhibit a particularly short half-life and difficult production. The development of an approach allowing simplified development, delivery, and in vivo production would be an important advance. Here we describe the development of a designed synthetic DNA plasmid, which we optimized to permit high expression of an anti-HER2 antibody (HER2dMAb) and delivered it into animals through adaptive electroporation. HER2dMAb was efficiently expressed in vitro and in vivo, reaching levels of 50 µg/ml in mouse sera. Mechanistically, HER2dMAb blocked HER2 signaling and induced antibody-dependent cytotoxicity. HER2dMAb delayed tumor progression for HER2-expressing ovarian and breast cancer models. We next used the HER2dMAb single-chain variable fragment portion to engineer a DNA-encoded BiTE (DBiTE). This HER2DBiTE was expressed in vivo for approximately 4 months after a single administration. The HER2DBiTE was highly cytolytic and delayed cancer progression in mice. These studies illustrate an approach to generate DBiTEs in vivo, which represent promising immunotherapies for HER2+ tumors, including ovarian and potentially other cancers.


Subject(s)
Antibodies, Bispecific/administration & dosage , Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents, Immunological/administration & dosage , Drug Delivery Systems/methods , Neoplasms/drug therapy , Animals , Antibodies, Bispecific/genetics , Antibodies, Monoclonal/genetics , Cell Line, Tumor , Electroporation/methods , Female , Humans , Male , Mice , Neoplasms/immunology , Neoplasms/pathology , Plasmids/administration & dosage , Plasmids/genetics , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/immunology , Receptor, ErbB-2/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Xenograft Model Antitumor Assays
15.
J Infect Dis ; 219(7): 1146-1150, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30476132

ABSTRACT

We recently developed anti-OspA human immunoglobulin G1 monoclonal antibodies (HuMAbs) that are effective in preventing Borrelia transmission from ticks in a murine model. Here, we investigated a novel approach of DNA-mediated gene transfer of HuMAbs that provide protection against Lyme disease. Plasmid DNA-encoded anti-OspA HuMAbs inoculated in mice achieved a serum antibody concentration of >6 µg/mL. Among mice injected with DNA-encoded monoclonal antibodies, 75%-77% were protected against an acute challenge by Borrelia-infected ticks. Our results represent the first demonstration of employing DNA transfer as a delivery system for antibodies that block transmission of Borrelia in animal models.


Subject(s)
Antibodies, Monoclonal, Humanized/immunology , Antigens, Surface/immunology , Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/immunology , DNA, Bacterial/immunology , Lipoproteins/immunology , Lyme Disease/transmission , Animals , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/therapeutic use , Antigens, Surface/genetics , Bacterial Outer Membrane Proteins/genetics , Bacterial Vaccines/genetics , Borrelia burgdorferi , Female , HEK293 Cells , Humans , Lipoproteins/genetics , Lyme Disease/prevention & control , Mice , Mice, Inbred C3H , Mice, SCID , Plasmids/immunology , Ticks , Transfection
16.
J Infect Dis ; 219(4): 544-555, 2019 01 29.
Article in English | MEDLINE | ID: mdl-30304515

ABSTRACT

Background: There remains an important need for prophylactic anti-Ebola virus vaccine candidates that elicit long-lasting immune responses and can be delivered to vulnerable populations that are unable to receive live-attenuated or viral vector vaccines. Methods: We designed novel synthetic anti-Ebola virus glycoprotein (EBOV-GP) DNA vaccines as a strategy to expand protective breadth against diverse EBOV strains and evaluated the impact of vaccine dosing and route of administration on protection against lethal EBOV-Makona challenge in cynomolgus macaques. Long-term immunogenicity was monitored in nonhuman primates for >1 year, followed by a 12-month boost. Results: Multiple-injection regimens of the EBOV-GP DNA vaccine, delivered by intramuscular administration followed by electroporation, were 100% protective against lethal EBOV-Makona challenge. Impressively, 2 injections of a simple, more tolerable, and dose-sparing intradermal administration followed by electroporation generated strong immunogenicity and was 100% protective against lethal challenge. In parallel, we observed that EBOV-GP DNA vaccination induced long-term immune responses in macaques that were detectable for at least 1 year after final vaccination and generated a strong recall response after the final boost. Conclusions: These data support that this simple intradermal-administered, serology-independent approach is likely important for additional study towards the goal of induction of anti-EBOV immunity in multiple at-risk populations.


Subject(s)
Ebola Vaccines/immunology , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/prevention & control , Vaccines, DNA/immunology , Animals , Disease Models, Animal , Ebola Vaccines/administration & dosage , Female , Injections, Intramuscular , Macaca fascicularis , Male , Vaccines, DNA/administration & dosage
17.
Cell Rep ; 25(7): 1982-1993.e4, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30428362

ABSTRACT

Synthetically engineered DNA-encoded monoclonal antibodies (DMAbs) are an in vivo platform for evaluation and delivery of human mAb to control against infectious disease. Here, we engineer DMAbs encoding potent anti-Zaire ebolavirus (EBOV) glycoprotein (GP) mAbs isolated from Ebola virus disease survivors. We demonstrate the development of a human IgG1 DMAb platform for in vivo EBOV-GP mAb delivery and evaluation in a mouse model. Using this approach, we show that DMAb-11 and DMAb-34 exhibit functional and molecular profiles comparable to recombinant mAb, have a wide window of expression, and provide rapid protection against lethal mouse-adapted EBOV challenge. The DMAb platform represents a simple, rapid, and reproducible approach for evaluating the activity of mAb during clinical development. DMAbs have the potential to be a mAb delivery system, which may be advantageous for protection against highly pathogenic infectious diseases, like EBOV, in resource-limited and other challenging settings.


Subject(s)
Antibodies, Monoclonal/immunology , DNA/administration & dosage , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/prevention & control , Animals , Disease Models, Animal , Epitope Mapping , Epitopes/immunology , Female , Glycoproteins/immunology , HEK293 Cells , Hemorrhagic Fever, Ebola/virology , Humans , Mice, Inbred BALB C , Muscles/metabolism , Mutagenesis , Recombinant Proteins/metabolism
18.
Cancer Res ; 78(22): 6363-6370, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30287678

ABSTRACT

Antibody-based immune therapies targeting the T-cell checkpoint molecules CTLA-4 and PD-1 have affected cancer therapy. However, this immune therapy requires complex manufacturing and frequent dosing, limiting the global use of this treatment. Here, we focused on the development of a DNA-encoded monoclonal antibody (DMAb) approach for delivery of anti-CTLA-4 monoclonal antibodies in vivo With this technology, engineered and formulated DMAb plasmids encoding IgG inserts were directly injected into muscle and delivered intracellularly by electroporation, leading to in vivo expression and secretion of the encoded IgG. DMAb expression from a single dose can continue for several months without the need for repeated administration. Delivery of an optimized DMAb encoding anti-mouse CTLA-4 IgG resulted in high serum levels of the antibody as well as tumor regression in Sa1N and CT26 tumor models. DNA-delivery of the anti-human CTLA-4 antibodies ipilimumab and tremelimumab in mice achieved potent peak levels of approximately 85 and 58 µg/mL, respectively. These DMAb exhibited prolonged expression, with maintenance of serum levels at or above 15 µg/mL for over a year. Anti-human CTLA-4 DMAbs produced in vivo bound to human CTLA-4 protein expressed on stimulated human peripheral blood mononuclear cells and induced T-cell activation in a functional assay ex vivo In summary, direct in vivo expression of DMAb encoding checkpoint inhibitors serves as a novel tool for immunotherapy that could significantly improve availability and provide broader access to such therapies.Significance: DNA-encoded monoclonal antibodies represent a novel technology for delivery and expression of immune checkpoint blockade antibodies, thus expanding patient access to, and possible clinical applications of, these therapies. Cancer Res; 78(22); 6363-70. ©2018 AACR.


Subject(s)
Antibodies, Monoclonal/chemistry , CTLA-4 Antigen/immunology , DNA/chemistry , Neoplasms/immunology , Neoplasms/therapy , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , CTLA-4 Antigen/antagonists & inhibitors , Cell Line, Tumor , HEK293 Cells , Humans , Immunoglobulin G/chemistry , Immunotherapy , Inhibitory Concentration 50 , Ipilimumab/pharmacology , Leukocytes, Mononuclear/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Transplantation , Plasmids/metabolism , T-Lymphocytes/metabolism
19.
EBioMedicine ; 35: 97-105, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30174283

ABSTRACT

BACKGROUND: Despite vigorous and ongoing efforts, active immunizations have yet to induce broadly neutralizing antibodies (bNAbs) against HIV-1. An alternative approach is to achieve prophylaxis with long-term expression of potent biologic HIV-1 inhibitors with Adeno-associated Virus (AAV), which could however be limited by hosts' humoral and cellular responses. An approach that facilitates in vivo production of these complex molecules independent of viral-vectored delivery will be a major advantage. METHODS: We used synthetic DNA and electroporation (DNA/EP) to deliver an anti-HIV-1 immunoadhesin eCD4-Ig in vivo. In addition, we engineered a TPST2 enzyme variant (IgE-TPST2), characterized its intracellular trafficking patterns and determined its ability to post-translationally sulfate eCD4-Ig in vivo. FINDINGS: With a single round of DNA injection, a peak expression level of 80-100µg/mL was observed in mice 14 days post injection (d.p.i). The engineered IgE-TPST2 enzyme trafficked efficiently to the Trans-Golgi Network (TGN). Co-administrating low dose of plasmid IgE-TPST2 with plasmid eCD4-Ig enhanced the potency of eCD4-Ig by three-fold in the ex vivo neutralization assay against the global panel of HIV-1 pseudoviruses. INTERPRETATION: This work provides a proof-of-concept for delivering anti-HIV-1 immunoadhesins by advanced nucleic acid technology and modulating protein functions in vivo with targeted enzyme-mediated post-translational modifications. FUNDING: This work is supported by NIH IPCAVD Grant U19 Al109646-04, Martin Delaney Collaboration for HIV Cure Research and W.W. Smith Charitable Trust.


Subject(s)
Antibodies, Neutralizing/metabolism , CD4 Immunoadhesins/metabolism , DNA/metabolism , Electroporation/methods , HIV Antibodies/metabolism , Immunoglobulin E/metabolism , Sulfates/metabolism , Animals , Female , HEK293 Cells , Humans , Mice, Inbred BALB C , Subcellular Fractions/metabolism , Transfection
20.
Mol Ther ; 26(2): 435-445, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29249395

ABSTRACT

Immune checkpoint blockade antibodies are setting a new standard of care for cancer patients. It is therefore important to assess any new immune-based therapies in the context of immune checkpoint blockade. Here, we evaluate the impact of combining a synthetic consensus TERT DNA vaccine that has improved capacity to break tolerance with immune checkpoint inhibitors. We observed that blockade of CTLA-4 or, to a lesser extent, PD-1 synergized with TERT vaccine, generating more robust anti-tumor activity compared to checkpoint alone or vaccine alone. Despite this anti-tumor synergy, none of these immune checkpoint therapies showed improvement in TERT antigen-specific immune responses in tumor-bearing mice. αCTLA-4 therapy enhanced the frequency of T-bet+/CD44+ effector CD8+ T cells within the tumor and decreased the frequency of regulatory T cells within the tumor, but not in peripheral blood. CTLA-4 blockade synergized more than Treg depletion with TERT DNA vaccine, suggesting that the effect of CTLA-4 blockade is more likely due to the expansion of effector T cells in the tumor rather than a reduction in the frequency of Tregs. These results suggest that immune checkpoint inhibitors function to alter the immune regulatory environment to synergize with DNA vaccines, rather than boosting antigen-specific responses at the site of vaccination.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Cancer Vaccines/immunology , Neoplasms/genetics , Neoplasms/immunology , Telomerase/immunology , Vaccines, DNA/immunology , Animals , Biomarkers, Tumor , CTLA-4 Antigen/antagonists & inhibitors , Cancer Vaccines/genetics , Cell Line, Tumor , Combined Modality Therapy , Disease Models, Animal , Female , Humans , Immunotherapy , Mice , Neoplasms/pathology , Neoplasms/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Telomerase/genetics , Vaccines, DNA/genetics , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...