Network


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

Hotspot


Dive into the research topics where Florian Klein is active.

Publication


Featured researches published by Florian Klein.


Science | 2011

Sequence and Structural Convergence of Broad and Potent HIV Antibodies That Mimic CD4 Binding

Johannes F. Scheid; Hugo Mouquet; Beatrix Ueberheide; Ron Diskin; Florian Klein; Thiago Y. Oliveira; John Pietzsch; David Fenyö; Alexander Abadir; Klara Velinzon; Arlene Hurley; Sunnie Myung; Farid Boulad; Pascal Poignard; Dennis R. Burton; Florencia Pereyra; David D. Ho; Bruce D. Walker; Michael S. Seaman; Pamela J. Bjorkman; Brian T. Chait; Michel C. Nussenzweig

Anti-HIV broadly neutralizing antibodies with similar specificities and modes of binding were found in multiple HIV-infected individuals. Passive transfer of broadly neutralizing HIV antibodies can prevent infection, which suggests that vaccines that elicit such antibodies would be protective. Thus far, however, few broadly neutralizing HIV antibodies that occur naturally have been characterized. To determine whether these antibodies are part of a larger group of related molecules, we cloned 576 new HIV antibodies from four unrelated individuals. All four individuals produced expanded clones of potent broadly neutralizing CD4-binding-site antibodies that mimic binding to CD4. Despite extensive hypermutation, the new antibodies shared a consensus sequence of 68 immunoglobulin H (IgH) chain amino acids and arise independently from two related IgH genes. Comparison of the crystal structure of one of the antibodies to the broadly neutralizing antibody VRC01 revealed conservation of the contacts to the HIV spike.


Nature | 2013

Therapeutic efficacy of potent neutralizing HIV-1-specific monoclonal antibodies in SHIV-infected rhesus monkeys

Dan H. Barouch; James B. Whitney; Brian Moldt; Florian Klein; Thiago Y. Oliveira; Jinyan Liu; Kathryn E. Stephenson; Hui-Wen Chang; Karthik Shekhar; Sanjana Gupta; Joseph P. Nkolola; Michael S. Seaman; Kaitlin M. Smith; Erica N. Borducchi; Crystal Cabral; Jeffrey Y. Smith; Stephen Blackmore; Srisowmya Sanisetty; James R. Perry; Matthew Beck; Mark G. Lewis; William Rinaldi; Arup K. Chakraborty; Pascal Poignard; Michel C. Nussenzweig; Dennis R. Burton

Human immunodeficiency virus type 1 (HIV-1)-specific monoclonal antibodies with extraordinary potency and breadth have recently been described. In humanized mice, combinations of monoclonal antibodies have been shown to suppress viraemia, but the therapeutic potential of these monoclonal antibodies has not yet been evaluated in primates with an intact immune system. Here we show that administration of a cocktail of HIV-1-specific monoclonal antibodies, as well as the single glycan-dependent monoclonal antibody PGT121, resulted in a rapid and precipitous decline of plasma viraemia to undetectable levels in rhesus monkeys chronically infected with the pathogenic simian–human immunodeficiency virus SHIV-SF162P3. A single monoclonal antibody infusion afforded up to a 3.1 log decline of plasma viral RNA in 7 days and also reduced proviral DNA in peripheral blood, gastrointestinal mucosa and lymph nodes without the development of viral resistance. Moreover, after monoclonal antibody administration, host Gag-specific T-lymphocyte responses showed improved functionality. Virus rebounded in most animals after a median of 56 days when serum monoclonal antibody titres had declined to undetectable levels, although, notably, a subset of animals maintained long-term virological control in the absence of further monoclonal antibody infusions. These data demonstrate a profound therapeutic effect of potent neutralizing HIV-1-specific monoclonal antibodies in SHIV-infected rhesus monkeys as well as an impact on host immune responses. Our findings strongly encourage the investigation of monoclonal antibody therapy for HIV-1 in humans.


Nature | 2012

HIV therapy by a combination of broadly neutralizing antibodies in humanized mice

Florian Klein; Ariel Halper-Stromberg; Joshua A. Horwitz; Henning Gruell; Johannes F. Scheid; Stylianos Bournazos; Hugo Mouquet; Linda Spatz; Ron Diskin; Alexander Abadir; Trinity Zang; Marcus Dorner; Eva Billerbeck; Rachael N. Labitt; Christian Gaebler; Paola M. Marcovecchio; Reha-Baris Incesu; Thomas R. Eisenreich; Paul D. Bieniasz; Michael S. Seaman; Pamela J. Bjorkman; Jeffrey V. Ravetch; Alexander Ploss; Michel C. Nussenzweig

Human antibodies to human immunodeficiency virus-1 (HIV-1) can neutralize a broad range of viral isolates in vitro and protect non-human primates against infection. Previous work showed that antibodies exert selective pressure on the virus but escape variants emerge within a short period of time. However, these experiments were performed before the recent discovery of more potent anti-HIV-1 antibodies and their improvement by structure-based design. Here we re-examine passive antibody transfer as a therapeutic modality in HIV-1-infected humanized mice. Although HIV-1 can escape from antibody monotherapy, combinations of broadly neutralizing antibodies can effectively control HIV-1 infection and suppress viral load to levels below detection. Moreover, in contrast to antiretroviral therapy, the longer half-life of antibodies led to control of viraemia for an average of 60 days after cessation of therapy. Thus, combinations of potent monoclonal antibodies can effectively control HIV-1 replication in humanized mice, and should be re-examined as a therapeutic modality in HIV-1-infected individuals.


Science | 2013

Antibodies in HIV-1 Vaccine Development and Therapy

Florian Klein; Hugo Mouquet; Pia Dosenovic; Johannes F. Scheid; Louise Scharf; Michel C. Nussenzweig

Despite 30 years of study, there is no HIV-1 vaccine and, until recently, there was little hope for a protective immunization. Renewed optimism in this area of research comes in part from the results of a recent vaccine trial and the use of single-cell antibody-cloning techniques that uncovered naturally arising, broad and potent HIV-1–neutralizing antibodies (bNAbs). These antibodies can protect against infection and suppress established HIV-1 infection in animal models. The finding that these antibodies develop in a fraction of infected individuals supports the idea that new approaches to vaccination might be developed by adapting the natural immune strategies or by structure-based immunogen design. Moreover, the success of passive immunotherapy in small-animal models suggests that bNAbs may become a valuable addition to the armamentarium of drugs that work against HIV-1.


Nature | 2013

Antibody-mediated immunotherapy of macaques chronically infected with SHIV suppresses viraemia

Masashi Shingai; Yoshiaki Nishimura; Florian Klein; Hugo Mouquet; Olivia K. Donau; Ronald J. Plishka; Alicia Buckler-White; Michael S. Seaman; Michael Piatak; Jeffrey D. Lifson; Dimiter S. Dimitrov; Michel C. Nussenzweig; Malcolm A. Martin

Neutralizing antibodies can confer immunity to primate lentiviruses by blocking infection in macaque models of AIDS. However, earlier studies of anti-human immunodeficiency virus type 1 (HIV-1) neutralizing antibodies administered to infected individuals or humanized mice reported poor control of virus replication and the rapid emergence of resistant variants. A new generation of anti-HIV-1 monoclonal antibodies, possessing extraordinary potency and breadth of neutralizing activity, has recently been isolated from infected individuals. These neutralizing antibodies target different regions of the HIV-1 envelope glycoprotein including the CD4-binding site, glycans located in the V1/V2, V3 and V4 regions, and the membrane proximal external region of gp41 (refs 9, 10, 11, 12, 13, 14). Here we have examined two of the new antibodies, directed to the CD4-binding site and the V3 region (3BNC117 and 10-1074, respectively), for their ability to block infection and suppress viraemia in macaques infected with the R5 tropic simian–human immunodeficiency virus (SHIV)-AD8, which emulates many of the pathogenic and immunogenic properties of HIV-1 during infections of rhesus macaques. Either antibody alone can potently block virus acquisition. When administered individually to recently infected macaques, the 10-1074 antibody caused a rapid decline in virus load to undetectable levels for 4–7 days, followed by virus rebound during which neutralization-resistant variants became detectable. When administered together, a single treatment rapidly suppressed plasma viraemia for 3–5 weeks in some long-term chronically SHIV-infected animals with low CD4+ T-cell levels. A second cycle of anti-HIV-1 monoclonal antibody therapy, administered to two previously treated animals, successfully controlled virus rebound. These results indicate that immunotherapy or a combination of immunotherapy plus conventional antiretroviral drugs might be useful as a treatment for chronically HIV-1-infected individuals experiencing immune dysfunction.


Science | 2011

Increasing the potency and breadth of an HIV antibody by using structure-based rational design.

Ron Diskin; Johannes F. Scheid; Paola M. Marcovecchio; Anthony P. West; Florian Klein; Han Gao; Priyanthi N. P. Gnanapragasam; Alexander Abadir; Michael S. Seaman; Michel C. Nussenzweig; Pamela J. Bjorkman

Structural analysis of an HIV antibody reveals residues important for neutralization breadth and potency. Antibodies against the CD4 binding site (CD4bs) on the HIV-1 spike protein gp120 can show exceptional potency and breadth. We determined structures of NIH45-46, a more potent clonal variant of VRC01, alone and bound to gp120. Comparisons with VRC01-gp120 revealed that a four-residue insertion in heavy chain complementarity–determining region 3 (CDRH3) contributed to increased interaction between NIH45-46 and the gp120 inner domain, which correlated with enhanced neutralization. We used structure-based design to create NIH45-46G54W, a single substitution in CDRH2 that increases contact with the gp120 bridging sheet and improves breadth and potency, critical properties for potential clinical use, by an order of magnitude. Together with the NIH45-46–gp120 structure, these results indicate that gp120 inner domain and bridging sheet residues should be included in immunogens to elicit CD4bs antibodies.


Cell | 2014

Structural Insights on the Role of Antibodies in HIV-1 Vaccine and Therapy

Anthony P. West; Louise Scharf; Johannes F. Scheid; Florian Klein; Pamela J. Bjorkman; Michel C. Nussenzweig

Despite 30 years of effort, there is no effective vaccine for HIV-1. However, antibodies can prevent HIV-1 infection in humanized mice and macaques when passively transferred. New single-cell-based methods have uncovered many broad and potent donor-derived antibodies, and structural studies have revealed the molecular bases for their activities. The new data suggest why such antibodies are difficult to elicit and inform HIV-1 vaccine development efforts. In addition to protecting against infection, the newly identified antibodies can suppress active infections in mice and macaques, suggesting they could be valuable additions to anti-HIV-1 therapies and to strategies to eradicate HIV-1 infection.


The FASEB Journal | 2003

Regulation of the multidrug resistance transporter P-glycoprotein in multicellular tumor spheroids by hypoxia-inducible factor (HIF-1) and reactive oxygen species

Maria Wartenberg; Frederike C. Ling; Markus Müschen; Florian Klein; H. Acker; Max Gassmann; Kerstin Petrat; Volker Pütz; Jürgen Hescheler; Heinrich Sauer

Hypoxia in tumors is generally associated with chemoresistance and radioresistance. However, the correlation between the heterodimeric hypoxia‐inducible factor‐1 (HIF‐1) and the multidrug resistance transporter P‐glycoprotein (P‐gp) has not been investigated. Herein, we demonstrate that with increasing size of DU‐145 prostate multicellular tumor spheroids the pericellular oxygen pressure and the generation of reactive oxygen species decreased, whereas the α‐subunit of HIF‐1 (HIF‐1α) and P‐gp were up‐regulated. Furthermore, P‐gp was up‐regulated under experimental physiological hypoxia and chemical hypoxia induced by either cobalt chloride or desferrioxamine. The pro‐oxidants H2O2 and buthionine sulfoximine down‐regulated HIF‐1α and P‐gp, whereas up‐regulation was achieved with the radical scavengers dehydroascorbate, N‐acetylcysteine, and vitamin E. The correlation of HIF‐1α and P‐gp expression was validated by the use of hepatoma tumor spheroids that were either wild type (Hepa1) or mutant (Hepa1C4) for aryl hydrocarbon receptor nuclear translocator (ARNT), i.e., HIF‐1β. Chemical hypoxia robustly increased HIF‐1α as well as P‐gp expression in Hepa1 tumor spheroids, whereas no changes were observed in Hepa1C4 spheroids. Hence, our data demonstrate that expression of P‐gp in multicellular tumor spheroids is under the control of HIF‐1.


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

HIV-1 suppression and durable control by combining single broadly neutralizing antibodies and antiretroviral drugs in humanized mice

Joshua A. Horwitz; Ariel Halper-Stromberg; Hugo Mouquet; Alexander D. Gitlin; Anna Tretiakova; Thomas R. Eisenreich; Marine Malbec; Sophia Gravemann; Eva Billerbeck; Marcus Dorner; Hildegard Büning; Olivier Schwartz; Elena Knops; Rolf Kaiser; Michael S. Seaman; James M. Wilson; Charles M. Rice; Alexander Ploss; Pamela J. Bjorkman; Florian Klein; Michel C. Nussenzweig

Significance Treatment of HIV-1 infection in humans is achieved using combinations of highly effective antiretroviral therapy (ART) drugs to potently suppress viral replication and prevent the emergence of drug-resistant viruses. However, ART drugs must be taken indefinitely owing to rapid return of viremia upon termination of treatment. Highly potent broadly neutralizing antibodies (bNAbs) present a new potential therapeutic modality in the treatment of HIV-1 infection. Because of their comparatively longer half-lives relative to ART drugs and their ability to eliminate infected cells, bNAbs may alleviate some aspects of the lifelong treatment adherence burden of ART. Here we show that lowering the initial viral load with ART enables single bNAbs to effectively control an established HIV-1 infection in humanized mice. Effective control of HIV-1 infection in humans is achieved using combinations of antiretroviral therapy (ART) drugs. In humanized mice (hu-mice), control of viremia can be achieved using either ART or by immunotherapy using combinations of broadly neutralizing antibodies (bNAbs). Here we show that treatment of HIV-1–infected hu-mice with a combination of three highly potent bNAbs not only resulted in complete viremic control but also led to a reduction in cell-associated HIV-1 DNA. Moreover, lowering the initial viral load by coadministration of ART and immunotherapy enabled prolonged viremic control by a single bNAb after ART was withdrawn. Similarly, a single injection of adeno-associated virus directing expression of one bNAb produced durable viremic control after ART was terminated. We conclude that immunotherapy reduces plasma viral load and cell-associated HIV-1 DNA and that decreasing the initial viral load enables single bNAbs to control viremia in hu-mice.


Journal of Experimental Medicine | 2014

Passive transfer of modest titers of potent and broadly neutralizing anti-HIV monoclonal antibodies block SHIV infection in macaques

Masashi Shingai; Olivia K. Donau; Ronald J. Plishka; Alicia Buckler-White; John R. Mascola; Gary J. Nabel; Martha Nason; David C. Montefiori; Brian Moldt; Pascal Poignard; Ron Diskin; Pamela J. Bjorkman; Michael A. Eckhaus; Florian Klein; Hugo Mouquet; Julio C. C. Lorenzi; Anna Gazumyan; Dennis R. Burton; Michel C. Nussenzweig; Malcolm A. Martin; Yoshiaki Nishimura

Five potent and broadly anti-HIV neutralizing monoclonal antibodies are able to block infection by two different SHIVs in monkeys. The authors show that antibodies targeting the outer glycan coat were the most effective and determined that titers of roughly 1:100 protected half the animals.

Collaboration


Dive into the Florian Klein's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael S. Seaman

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Pamela J. Bjorkman

California Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anthony P. West

California Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ron Diskin

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge