Network


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

Hotspot


Dive into the research topics where Vladimir Liska is active.

Publication


Featured researches published by Vladimir Liska.


Nature Medicine | 2000

Human neutralizing monoclonal antibodies of the IgG1 subtype protect against mucosal simian-human immunodeficiency virus infection.

Timothy W. Baba; Vladimir Liska; Regina Hofmann-Lehmann; Josef Vlasak; Weidong Xu; Seyoum Ayehunie; Lisa A. Cavacini; Marshall R. Posner; Hermann Katinger; Gabriela Stiegler; Bruce J. Bernacky; Tahir A. Rizvi; Russell D. Schmidt; Lori R. Hill; Michale E. Keeling; Yichen Lu; Joel E. Wright; Ting Chao Chou; Ruth M. Ruprecht

Although maternal human immunodeficiency virus type 1 (HIV-1) transmission occurs during gestation, intrapartum and postpartum (by breast-feeding), 50–70% of all infected children seem to acquire HIV-1 shortly before or during delivery. Epidemiological evidence indicates that mucosal exposure is an important aspect of intrapartum HIV transmission. A simian immunodeficiency virus (SIV) macaque model has been developed that mimics the mucosal exposure that can occur during intrapartum HIV-1 transmission. To develop immunoprophylaxis against intrapartum HIV-1 transmission, we used SHIV–vpu+ (refs. 5,6), a chimeric simian–human virus that encodes the env gene of HIV-IIIB. Several combinations of human monoclonal antibodies against HIV-1 have been identified that neutralize SHIV–vpu+ completely in vitro through synergistic interaction. Here, we treated four pregnant macaques with a triple combination of the human IgG1 monoclonal antibodies F105, 2G12 and 2F5. All four macaques were protected against intravenous SHIV–vpu+ challenge after delivery. The infants received monoclonal antibodies after birth and were challenged orally with SHIV–vpu+ shortly thereafter. We found no evidence of infection in any infant during 6 months of follow-up. This demonstrates that IgG1 monoclonal antibodies protect against mucosal lentivirus challenge in neonates. We conclude that epitopes recognized by the three monoclonal antibodies are important determinants for achieving substantial protection, thus providing a rational basis for AIDS vaccine development.


Nature Medicine | 1999

Live attenuated, multiply deleted simian immunodeficiency virus causes AIDS in infant and adult macaques.

Timothy W. Baba; Vladimir Liska; Anis H Khimani; Nancy B. Ray; Peter J. Dailey; Dominique G. Penninck; Rod Bronson; Michael F. Greene; Harold M. McClure; Louis N. Martin; Ruth M. Ruprecht

A substantial risk in using live attenuated, multiply deleted viruses as vaccines against AIDS is their potential to induce AIDS. A mutant of the simian immunodeficiency virus (SIV) with large deletions in nef and vpr and in the negative regulatory element induced AIDS in six of eight infant macaques vaccinated orally or intravenously. Early signs of immune dysfunction were seen in the remaining two offspring. Prolonged follow–up of sixteen vaccinated adult macaques also showed resurgence of chronic viremia in four animals: two of these developed early signs of disease and one died of AIDS. We conclude that this multiply deleted SIV is pathogenic and that human AIDS vaccines built on similar prototypes may cause AIDS.


AIDS Research and Human Retroviruses | 2000

Sensitive and robust one-tube real-time reverse transcriptase-polymerase chain reaction to quantify SIV RNA load: comparison of one- versus two-enzyme systems.

Regina Hofmann-Lehmann; Ryan K. Swenerton; Vladimir Liska; Christian M. Leutenegger; Hans Lutz; Harold M. McClure; Ruth M. Ruprecht

Plasma viral RNA load is a key parameter in disease progression of lentiviral infections. To measure simian immunodeficiency virus (SIV) RNA loads, we have established a quantitative one-tube assay based on TaqMan chemistry. This real-time reverse transcriptase-polymerase chain reaction (RT-PCR) has advantages compared with previous methods, such as higher sensitivity, shorter time consumption, and low risk of cross-contamination. The sensitivity of the assay was optimized by comparing different enzyme systems. The one-enzyme protocol using rTth DNA polymerase was superior to two assays employing two enzymes. It detects 100% of the samples containing four copies of RNA transcript and allows quantification of viral RNA loads over an 8-log unit dynamic range. As few as 50 copies per milliliter of plasma can be detected within RNA extracted from 140 microl of plasma. This is especially relevant in studies employing neonatal macaques, from which only small volumes of blood can be sampled, and in studies in which low viral RNA loads are expected. Because of the use of rTth DNA polymerase, DNA contamination can be avoided by carryover prevention with uracil N-glycosylase (UNG). We demonstrate that for optimization of real-time PCR sensitivity, not only concentrations of different reagents but also different enzyme systems must be evaluated. Our assay facilitates and enhances the quantification of plasma RNA loads, a critical parameter for many studies, including evaluations of vaccine candidates or antiviral regimens.


Journal of Virology | 2001

Postnatal Passive Immunization of Neonatal Macaques with a Triple Combination of Human Monoclonal Antibodies against Oral Simian-Human Immunodeficiency Virus Challenge

Regina Hofmann-Lehmann; Josef Vlasak; Robert A. Rasmussen; Smith B; Timothy W. Baba; Vladimir Liska; Flavia Ferrantelli; David C. Montefiori; Harold M. McClure; Daniel C. Anderson; Bruce J. Bernacky; Tahir A. Rizvi; Russell D. Schmidt; Lori R. Hill; Michale E. Keeling; Hermann Katinger; Gabriela Stiegler; Lisa A. Cavacini; Marshall R. Posner; Ting-Chao Chou; Janet Andersen; Ruth M. Ruprecht

ABSTRACT To develop prophylaxis against mother-to-child human immunodeficiency virus (HIV) transmission, we established a simian-human immunodeficiency virus (SHIV) infection model in neonatal macaques that mimics intrapartum mucosal virus exposure (T. W. Baba et al., AIDS Res. Hum. Retroviruses 10:351–357, 1994). Using this model, neonates were protected from mucosal SHIV-vpu+challenge by pre- and postnatal treatment with a combination of three human neutralizing monoclonal antibodies (MAbs), F105, 2G12, and 2F5 (Baba et al., Nat. Med. 6:200–206, 2000). In the present study, we used this MAb combination only postnatally, thereby significantly reducing the quantity of antibodies necessary and rendering their potential use in humans more practical. We protected two neonates with this regimen against oral SHIV-vpu+ challenge, while four untreated control animals became persistently infected. Thus, synergistic MAbs protect when used as immunoprophylaxis without the prenatal dose. We then determined in vitro the optimal MAb combination against the more pathogenic SHIV89.6P, a chimeric virus encodingenv of the primary HIV89.6. Remarkably, the most potent combination included IgG1b12, which alone does not neutralize SHIV89.6P. We administered the combination of MAbs IgG1b12, 2F5, and 2G12 postnatally to four neonates. One of the four infants remained uninfected after oral challenge with SHIV89.6P, and two infants had no or a delayed CD4+ T-cell decline. In contrast, all control animals had dramatic drops in their CD4+ T cells by 2 weeks postexposure. We conclude that our triple MAb combination partially protected against mucosal challenge with the highly pathogenic SHIV89.6P. Thus, combination immunoprophylaxis with passively administered synergistic human MAbs may play a role in the clinical prevention of mother-to-infant transmission of HIV type 1.


Science | 1996

Infection and AIDS in Adult Macaques After Nontraumatic Oral Exposure to Cell-Free SIV

Timothy W. Baba; Anita M. Trichel; Li An; Vladimir Liska; Louis N. Martin; Michael Murphey-Corb; Ruth M. Ruprecht

Unprotected receptive anal intercourse is a well-recognized risk factor for infection with human immunodeficiency virus-type 1 (HIV-1). Isolated human case reports have implicated HIV-1 transmission by oral-genital exposure. Adult macaques exposed nontraumatically to cell-free simian immunodeficiency virus (SIV) through the oral route became infected and developed acquired immunodeficiency syndrome (AIDS). The minimal virus dose needed to achieve systemic infection after oral exposure was 6000 times lower than the minimal dose required to achieve systemic infection after rectal exposure. Thus, unprotected receptive oral intercourse, even in the absence of mucosal lesions, should be added to the list of risk behaviors for HIV-1 transmission.


Transfusion Clinique Et Biologique | 2001

Protection of neonatal macaques against experimental SHIV infection by human neutralizing monoclonal antibodies

Ruth M. Ruprecht; Regina Hofmann-Lehmann; Smith-Franklin Ba; Robert A. Rasmussen; Vladimir Liska; Josef Vlasak; Weidong Xu; Timothy W. Baba; Agnès-Laurence Chenine; Lisa A. Cavacini; Marshall R. Posner; Hermann Katinger; Gabriela Stiegler; Bruce J. Bernacky; Tahir A. Rizvi; Russell D. Schmidt; Lori R. Hill; Michale E. Keeling; David C. Montefiori; Harold M. McClure

Neonatal macaques were completely protected against oral challenge with SHIV-vpu+, a simian-human immunodeficiency virus that encodes the envelope gene of a laboratory-adapted HIV strain, by pre- and post-natal treatment with a triple combination of human neutralizing monoclonal antibodies (mAbs). The mAbs were directed either against the CD4 binding site, a glycosylation-dependent gp120 epitope, or against a linear epitope on gp41. This triple combination was highly synergistic in vitro and neutralized primary HIV completely. Subsequently, oral challenge was performed with pathogenic SHIV89.6P, an animal-passaged variant of a chimeric virus that encodes the envelope gene of the primary, dual-tropic HIV89.6. Only post-natal treatment with a similar triple mAb combination was used. One out of 4 mAb-treated infants was completely protected from infection. In the other 3 treated animals, there was a tendency towards lower peak viral RNA loads compared with untreated controls. Two out of 4 mAb-treated infants maintained normal CD4+ T-cell numbers, in contrast to all controls that had steep declines at 2 weeks post-challenge. We conclude that the triple mAb combination significantly protected the neonates, even against mucosal challenge with pathogenic SHIV89.6P. Passively administered synergistic human mAbs may play a role in preventing mother-infant transmission of HIV, both against intrapartum transmission as well as against infection through breast milk. As passive immunization is a tool to assess correlates of immune protection, we conclude that the epitopes recognized by the mAbs in our combinations are important for AIDS vaccine development. Future passive immunization studies may reveal other important conserved epitopes.


The Journal of Infectious Diseases | 1999

Oral Transmission of Primate Lentiviruses

Ruth M. Ruprecht; Timothy W. Baba; Vladimir Liska; Nancy B. Ray; Louis N. Martin; Michael Murphey-Corb; Tahir A. Rizvi; Bruce J. Bernacky; Michale E. Bernacky; Harold M. McClure; Janet Andersen

Oral transmission of human immunodeficiency virus type 1 (HIV-1) is well documented in children who become infected postnatally through breast milk. In contrast, epidemiologic surveys have yielded conflicting data regarding oral HIV-1 transmission among adults, even though case reports have described seroconversion and the development of AIDS in adults whose only risk was oral-genital contact. To study oral virus transmission in primate models, we exposed rhesus macaques of various ages to cell-free simian immunodeficiency virus (SIV), including uncloned and molecularly cloned viruses. In neonates, viremia and AIDS developed after nontraumatic oral exposure to several SIV strains. Furthermore, chimeric simian human immunodeficiency viruses containing the HIV-1 envelope can also cross intact upper gastrointestinal mucosal surfaces in neonates. In adult macaques, infection and AIDS have resulted from well-controlled, nontraumatic, experimental oral exposure to different strains of SIV. These findings have implications for the risks of HIV-1 transmission during oral-genital contact.


Journal of Medical Primatology | 2001

Passive immunization against oral AIDS virus transmission: An approach to prevent mother‐to‐infant HIV‐1 transmission?

Regina Hofmann-Lehmann; Robert A. Rasmussen; Josef Vlasak; Smith B; Timothy W. Baba; Vladimir Liska; David C. Montefiori; Harold M. McClure; Daniel C. Anderson; Bruce J. Bernacky; Tahir A. Rizvi; Russell D. Schmidt; Lori R. Hill; Michale E. Keeling; Hermann Katinger; Gabriela Stiegler; Marshall R. Posner; Lisa A. Cavacini; Ting Chao Chou; Ruth M. Ruprecht

To develop immunoprophylaxis regimens against mother‐to‐child human immunodeficiency virus type 1 (HIV‐1) transmission, we established a simian–human immunodeficiency virus (SHIV) model in neonatal macaques that mimics intrapartum mucosal virus exposure (T.W. Baba, J. Koch, E.S. Mittler et al.: AIDS Res Hum Retroviruses 10:351–357, 1994). We protected four neonates from oral SHIV‐vpu+ challenge by ante‐ and postpartum treatment with a synergistic triple combination of immunoglobulin (Ig) G1 human anti‐HIV‐1 neutralizing monoclonal antibodies (mAbs) (T.W. Baba, V. Liska, R. Hofmann‐Lehmann et al.: Nature Med 6:200–206, 2000), which recognize the CD4‐binding site of Env, a glycosylation‐dependent gp120, or a linear gp41 epitope. Two neonates that received only postpartum mAbs were also protected from oral SHIV‐vpu+ challenge, indicating that postpartum treatment alone is sufficient. Next, we evaluated a similar mAb combination against SHIV89.6P, which encodes env of primary HIV89.6. One of four mAb‐treated neonates was protected from infection and two maintained normal CD4+ T‐cell counts. We conclude that the epitopes recognized by the three mAbs are important determinants for achieving protection. Combination immunoprophylaxis with synergistic mAbs seems promising to prevent maternal HIV‐1 transmission in humans.


Journal of Medical Primatology | 2002

DNA prime/protein boost vaccine strategy in neonatal macaques against simian human immunodeficiency virus

Robert A. Rasmussen; Regina Hofmann-Lehmann; David C. Montefiori; Pei-Lin Li; Vladimir Liska; Josef Vlasak; Timothy W. Baba; Jörn E. Schmitz; Marcelo J. Kuroda; Harriet L. Robinson; Harold M. McClure; Shan Lu; Shiu-Lok Hu; Tahir A. Rizvi; Ruth M. Ruprecht

Newborn macaques were vaccinated against a chimeric simian human immunodeficiency (SHIV) virus, SHIV‐vpu+, by DNA priming and boosting with homologous HIV‐1 gp160. Following SHIV‐vpu+ challenge, containment of infection was observed in 4 of 15 animals given DNA priming/protein boost vaccination and in three of four animals given gp160 boosts only. Rechallenge with homologous virus of six animals that contained the first challenge virus resulted in rapid viral clearance or low viral loads. Upon additional rechallenge with heterologous, pathogenic SHIV89.6P, four of these six animals maintained normal CD4+ T‐cell counts with no or limited SHIV89.6P infection. Our data suggest that humoral and cellular immune mechanisms may have contributed to the containment of SHIV89.6P; however, viral interference with SHIV‐vpu+ could also have played a role. Our results indicate that immunogenicity and efficacy of candidate AIDS vaccines are not affected when vaccination is initiated during infancy as compared with later in life.


AIDS Research and Human Retroviruses | 1999

Viremia and AIDS in rhesus macaques after intramuscular inoculation of plasmid DNA encoding full-length SIVmac239.

Vladimir Liska; Anis H Khimani; Regina Hofmann-Lehmann; Alice Ng Fink; Josef Vlasak; Ruth M. Ruprecht

We have succeeded in stably maintaining the entire genome of SIVmac239 as a plasmid clone. Supercoiled proviral plasmid DNA was inoculated intramuscularly into two adult rhesus macaques and into a neonate. All three animals became viremic and seroconverted. Viral kinetics were followed prospectively by quantitative competitive reverse transcriptase polymerase chain reaction (QC-RT-PCR), measurement of proviral DNA load in peripheral blood mononuclear cells (PBMCs) by PCR, and virus isolation by cocultivation. The infant developed high virus loads and succumbed to AIDS and SIV-associated nephropathy at 10 weeks postinoculation. Both adults are still living but have progressed to AIDS; one adult has also developed severe thrombocytopenia. We conclude that infection through intramuscular inoculation of cloned plasmid DNA encoding the entire proviral genome is reproducible and will provide a useful tool for studying viral pathogenesis.

Collaboration


Dive into the Vladimir Liska's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Harold M. McClure

Yerkes National Primate Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tahir A. Rizvi

United Arab Emirates University

View shared research outputs
Top Co-Authors

Avatar

Bruce J. Bernacky

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michale E. Keeling

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge