Network


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

Hotspot


Dive into the research topics where Helga Raab is active.

Publication


Featured researches published by Helga Raab.


Cell | 1990

Molecular cloning and expression of a receptor for human tumor necrosis factor

Thomas J. Schall; Martyn Lewis; Kerry J. Koller; Angela Lee; Glenn C. Rice; Grace H.W. Wong; Tetsuya Gatanaga; Gale A. Granger; Rigdon Lentz; Helga Raab; William J. Kohr; David V. Goeddel

A human tumor necrosis factor (TNF) binding protein from serum of cancer patients was purified to homogeneity and partially sequenced. Synthetic DNA probes based on amino acid sequence information were used to isolate cDNA clones encoding a receptor for TNF. The TNF receptor (TNF-R) is a 415 amino acid polypeptide with a single membrane-spanning region. The extracellular cysteine-rich domain of the TNF-R is homologous to the nerve growth factor receptor and the B cell activation protein Bp50. Human embryonic kidney cells transfected with a TNF-R expression vector specifically bind both 125I-labeled and biotinylated TNF-alpha. Unlabeled TNF-alpha and TNF-beta were equally effective at displacing the binding of labeled TNF-alpha to TNF-R expressing cells. Northern analysis indicates a single species of mRNA for the TNF-R in a variety of cell types. Therefore, the soluble TNF binding protein found in human serum is probably proteolytically derived from the TNF-R.


Science | 1992

Identification of Heregulin, a Specific Activator of p185erbB2

William E. Holmes; Mark X. Sliwkowski; Robert W. Akita; William J. Henzel; James Lee; John W. Park; Daniel G. Yansura; Nasrin Abadi; Helga Raab; Gail D. Lewis; H. Michael Shepard; Wun-Jing Kuang; William L. Wood; David V. Goeddel; Richard Vandlen

The proto-oncogene designated erbB2 or HER2 encodes a 185-kilodalton transmembrane tyrosine kinase (p185erbB2), whose overexpression has been correlated with a poor prognosis in several human malignancies. A 45-kilodalton protein heregulin-α (HRG-α) that specifically induced phosphorylation of p185erbB2 was purified from the conditioned medium of a human breast tumor cell line. Several complementary DNA clones encoding related HRGs were identified, all of which are similar to proteins in the epidermal growth factor family. Scatchard analysis of the binding of recombinant HRG to a breast tumor cell line expressing p185erbB2 showed a single high affinity binding site [dissociation constant (Kd) = 105 � 15 picomolar]. Heregulin transcripts were identified in several normal tissues and cancer cell lines. The HRGs may represent the natural ligands for p185erbB2.


Nature Biotechnology | 2008

Site-specific conjugation of a cytotoxic drug to an antibody improves the therapeutic index

Jagath R. Junutula; Helga Raab; Suzanna Clark; Sunil Bhakta; Douglas D. Leipold; Sylvia Weir; Yvonne Chen; Michelle Simpson; Siao Ping Tsai; Mark S. Dennis; Yanmei Lu; Y. Gloria Meng; Carl Ng; Jihong Yang; Chien C Lee; Eileen T. Duenas; Jeffrey Gorrell; Viswanatham Katta; Amy Kim; Kevin McDorman; Kelly Flagella; Rayna Venook; Sarajane Ross; Susan D. Spencer; Wai Lee Wong; Henry B. Lowman; Richard Vandlen; Mark X. Sliwkowski; Richard H. Scheller; Paul Polakis

Antibody-drug conjugates enhance the antitumor effects of antibodies and reduce adverse systemic effects of potent cytotoxic drugs. However, conventional drug conjugation strategies yield heterogenous conjugates with relatively narrow therapeutic index (maximum tolerated dose/curative dose). Using leads from our previously described phage display–based method to predict suitable conjugation sites, we engineered cysteine substitutions at positions on light and heavy chains that provide reactive thiol groups and do not perturb immunoglobulin folding and assembly, or alter antigen binding. When conjugated to monomethyl auristatin E, an antibody against the ovarian cancer antigen MUC16 is as efficacious as a conventional conjugate in mouse xenograft models. Moreover, it is tolerated at higher doses in rats and cynomolgus monkeys than the same conjugate prepared by conventional approaches. The favorable in vivo properties of the near-homogenous composition of this conjugate suggest that our strategy offers a general approach to retaining the antitumor efficacy of antibody-drug conjugates, while minimizing their systemic toxicity.


Nature Biotechnology | 2012

Conjugation site modulates the in vivo stability and therapeutic activity of antibody-drug conjugates

Ben-Quan Shen; Keyang Xu; Luna Liu; Helga Raab; Sunil Bhakta; Margaret Kenrick; Kathryn Parsons-Reponte; Janet Tien; Shang-Fan Yu; Elaine Mai; Dongwei Li; Jay Tibbitts; Jakub Baudys; Ola Saad; Suzie J. Scales; Paul J. Mcdonald; Philip E. Hass; Charles Eigenbrot; Trung Nguyen; Willy Solis; Reina N. Fuji; Kelly Flagella; Darshana Ramesh Patel; Susan D. Spencer; Leslie A. Khawli; Allen Ebens; Wai Lee Wong; Richard Vandlen; Surinder Kaur; Mark X. Sliwkowski

The reactive thiol in cysteine is used for coupling maleimide linkers in the generation of antibody conjugates. To assess the impact of the conjugation site, we engineered cysteines into a therapeutic HER2/neu antibody at three sites differing in solvent accessibility and local charge. The highly solvent-accessible site rapidly lost conjugated thiol-reactive linkers in plasma owing to maleimide exchange with reactive thiols in albumin, free cysteine or glutathione. In contrast, a partially accessible site with a positively charged environment promoted hydrolysis of the succinimide ring in the linker, thereby preventing this exchange reaction. The site with partial solvent-accessibility and neutral charge displayed both properties. In a mouse mammary tumor model, the stability and therapeutic activity of the antibody conjugate were affected positively by succinimide ring hydrolysis and negatively by maleimide exchange with thiol-reactive constituents in plasma. Thus, the chemical and structural dynamics of the conjugation site can influence antibody conjugate performance by modulating the stability of the antibody-linker interface.


mAbs | 2014

Site-specific antibody drug conjugates for cancer therapy

Siler Panowski; Sunil Bhakta; Helga Raab; Paul Polakis; Jagath R. Junutula

Antibody therapeutics have revolutionized the treatment of cancer over the past two decades. Antibodies that specifically bind tumor surface antigens can be effective therapeutics; however, many unmodified antibodies lack therapeutic activity. These antibodies can instead be applied successfully as guided missiles to deliver potent cytotoxic drugs in the form of antibody drug conjugates (ADCs). The success of ADCs is dependent on four factors—target antigen, antibody, linker, and payload. The field has made great progress in these areas, marked by the recent approval by the US Food and Drug Administration of two ADCs, brentuximab vedotin (Adcetris®) and ado-trastuzumab emtansine (Kadcyla®). However, the therapeutic window for many ADCs that are currently in pre-clinical or clinical development remains narrow and further improvements may be required to enhance the therapeutic potential of these ADCs. Production of ADCs is an area where improvement is needed because current methods yield heterogeneous mixtures that may include 0–8 drug species per antibody molecule. Site-specific conjugation has been recently shown to eliminate heterogeneity, improve conjugate stability, and increase the therapeutic window. Here, we review and describe various site-specific conjugation strategies that are currently used for the production of ADCs, including use of engineered cysteine residues, unnatural amino acids, and enzymatic conjugation through glycotransferases and transglutaminases. In addition, we also summarize differences among these methods and highlight critical considerations when building next-generation ADC therapeutics.


Clinical Cancer Research | 2010

Engineered Thio-Trastuzumab-DM1 Conjugate with an Improved Therapeutic Index to Target Human Epidermal Growth Factor Receptor 2–Positive Breast Cancer

Jagath R. Junutula; Kelly Flagella; Richard A. Graham; Kathryn Parsons; Edward Ha; Helga Raab; Sunil Bhakta; Trung Nguyen; Debra L. Dugger; Guangmin Li; Elaine Mai; Gail Lewis Phillips; Hajime Hiraragi; Reina N. Fuji; Jay Tibbitts; Richard Vandlen; Susan D. Spencer; Richard H. Scheller; Paul Polakis; Mark X. Sliwkowski

Purpose: Antibody drug conjugates (ADCs) combine the ideal properties of both antibodies and cytotoxic drugs by targeting potent drugs to the antigen-expressing tumor cells, thereby enhancing their antitumor activity. Successful ADC development for a given target antigen depends on optimization of antibody selection, linker stability, cytotoxic drug potency, and mode of linker-drug conjugation to the antibody. Here, we systematically examined the in vitro potency as well as in vivo preclinical efficacy and safety profiles of a heterogeneous preparation of conventional trastuzumab-mcc-DM1 (TMAb-mcc-DM1) ADC with that of a homogeneous engineered thio-trastuzumab-mpeo-DM1 (thioTMAb-mpeo-DM1) conjugate. Experimental Design and Results: To generate thioTMAb-mpeo-DM1, one drug maytansinoid 1 (DM1) molecule was conjugated to an engineered cysteine residue at Ala114 (Kabat numbering) on each trastuzumab-heavy chain, resulting in two DM1 molecules per antibody. ThioTMAb-mpeo-DM1 retained similar in vitro anti–cell proliferation activity and human epidermal growth factor receptor 2 (HER2) binding properties to that of the conventional ADC. Furthermore, it showed improved efficacy over the conventional ADC at DM1-equivalent doses (μg/m2) and retained efficacy at equivalent antibody doses (mg/kg). An improved safety profile of >2-fold was observed in a short-term target-independent rat safety study. In cynomolgus monkey safety studies, thioTMAb-mpeo-DM1 was tolerated at higher antibody doses (up to 48 mg/kg or 6,000 μg DM1/m2) compared with the conventional ADC that had dose-limiting toxicity at 30 mg/kg (6,000 μg DM1/m2). Conclusions: The engineered thioTMAb-mpeo-DM1 with broadened therapeutic index represents a promising antibody drug conjugate for future clinical development of HER2-positive targeted breast cancer therapies. Clin Cancer Res; 16(19); 4769–78. ©2010 AACR.


Cell | 1995

Reevaluation of the roles of protein S and gas6 as ligands for the receptor tyrosine kinase Rse/Tyro 3

Paul J. Godowski; Melanie R. Mark; Jian Chen; Michael D. Sadick; Helga Raab; R. Glenn Hammonds

Recently, Stitt et al. (1995) reported that protein S (PS), but not Gas6, is a potent ligand for the receptor tyrosine kinase known as Rse, Tyro 3, Brt, Sky, and Tif (hereafter referred to as Rse/Tyro 3). PS is an abundant serum protein previously characterized as an essential anticoagu-lant. Gas6, which was identified as a gene whose expression is increased by growth arrest, shares 43% amino acid identity and overall domain organization with PS (Manfio-letti et al., 1993). Stitt et al. (1995) based their conclusions on experiments describing interspecies interactions of bovine Gas6 with murine Rse/Tyro 3 (mRse/Tyro 3) and on interactions of bovine and human PS (hPS) with mRse/ Tyro 3. Consistent with the results of Stitt et al. (1995), we identified bovine PS as a ligand for human Rse/Tyro 3 (hRse/Tyro 3), and we also found that hPS can act as a ligand for mRse/Tyro 3. However, when we analyzed the more relevant intraspecies interactions, we obtained different results. We found that human Gas6 (hGas6), but not hPS, acted as a potent ligand for hRse/Tyro 3 (Figure 1). The hRse/Tyro 3 we have studied is very likely the true homolog of mRse/Tyro 3 since they share 90% amino acid identity and a similar expression pattern (Mark et al., 1994; Lai et al., 1994). To characterize the Rse/Tyro 3 ligand, we constructed soluble receptor proteins containing the extraceltular domain of either hRse/Tyro 3 or mRseFryro 3 fused to the Fc portion of human immunoglobulin G1 (hRse-lgG and mRse-lgG). A similar fusion protein (termed Tyro 3-Fc) was utilized by Stitt et al. (1995) to characterize the binding of bovine Gas6 and hPS to mRse/Tyro 3. We first determined whether hRse-lgG or mRse-lgG differed in its ability to bind to hPS or hGas6 containing an epitope tag that allows for side-by-side comparison of the binding properties of the two proteins. Either h Rse-lgG or mRse-lgG was incubated with conditioned medium containing putative ligands. Complexes were captured with protein A (specific for the IgG fusion protein) and visualized with an antibody specific for the epitope-tagged putative ligand. While hGas6 was bound by hRse-lgG, it was not efficiently bound by mRse-lgG (Figure 2). The reciprocal result was obtained in analysis of binding of hPS to hRse-lgG and mRse-lgG; hPS was bound by mRse-lgG but not by hRse-lgG. These results are consistent with an apparent difference in affinity of hPS for human as opposed …


Nature | 2015

Novel antibody–antibiotic conjugate eliminates intracellular S. aureus

Sophie M. Lehar; Thomas H. Pillow; Min Xu; Leanna Staben; Kimberly Kajihara; Richard Vandlen; Laura DePalatis; Helga Raab; Wouter L. W. Hazenbos; J. Hiroshi Morisaki; Janice Kim; Summer Park; Martine Darwish; Byoung-Chul Lee; Hilda Hernandez; Kelly M. Loyet; Patrick Lupardus; Rina Fong; Donghong Yan; Cecile Chalouni; Elizabeth Luis; Yana Khalfin; Emile Plise; Jonathan Cheong; Joseph P. Lyssikatos; Magnus Strandh; Klaus Koefoed; Peter S. Andersen; John A. Flygare; Man Wah Tan

Staphylococcus aureus is considered to be an extracellular pathogen. However, survival of S. aureus within host cells may provide a reservoir relatively protected from antibiotics, thus enabling long-term colonization of the host and explaining clinical failures and relapses after antibiotic therapy. Here we confirm that intracellular reservoirs of S. aureus in mice comprise a virulent subset of bacteria that can establish infection even in the presence of vancomycin, and we introduce a novel therapeutic that effectively kills intracellular S. aureus. This antibody–antibiotic conjugate consists of an anti-S. aureus antibody conjugated to a highly efficacious antibiotic that is activated only after it is released in the proteolytic environment of the phagolysosome. The antibody–antibiotic conjugate is superior to vancomycin for treatment of bacteraemia and provides direct evidence that intracellular S. aureus represents an important component of invasive infections.


Cancer Research | 2009

Antibody-Drug Conjugates for the Treatment of Non–Hodgkin's Lymphoma: Target and Linker-Drug Selection

Andrew Polson; Jill Calemine-Fenaux; Pamela Chan; Wesley Chang; Erin H. Christensen; Suzanna Clark; Frederic J. de Sauvage; Dan L. Eaton; Kristi Elkins; J. Michael Elliott; Gretchen Frantz; Reina N. Fuji; Alane Gray; Kristin Harden; Gladys Ingle; Noelyn M. Kljavin; Hartmut Koeppen; Christopher P. Nelson; Saileta Prabhu; Helga Raab; Sarajane Ross; Jean-Philippe Stephan; Suzie J. Scales; Susan D. Spencer; Richard Vandlen; Bernd Wranik; Shang-Fan Yu; Bing Zheng; Allen Ebens

Antibody-drug conjugates (ADC), potent cytotoxic drugs covalently linked to antibodies via chemical linkers, provide a means to increase the effectiveness of chemotherapy by targeting the drug to neoplastic cells while reducing side effects. Here, we systematically examine the potential targets and linker-drug combinations that could provide an optimal ADC for the treatment for non-Hodgkins lymphoma. We identified seven antigens (CD19, CD20, CD21, CD22, CD72, CD79b, and CD180) for potential treatment of non-Hodgkins lymphoma with ADCs. ADCs with cleavable linkers mediated in vivo efficacy via all these targets; ADCs with uncleavable linkers were only effective when targeted to CD22 and CD79b. In target-independent safety studies in rats, the uncleavable linker ADCs showed reduced toxicity, presumably due to the reduced release of free drug or other toxic metabolites into the circulation. Thus, our data suggest that ADCs with cleavable linkers work on a broad range of targets, and for specific targets, ADCs with uncleavable linkers provide a promising opportunity to improve the therapeutic window for ADCs in humans.


Journal of Biological Chemistry | 2008

Comprehensive Analysis of the Factors Contributing to the Stability and Solubility of Autonomous Human VH Domains.

Pierre A. Barthelemy; Helga Raab; Brent A. Appleton; Christopher J. Bond; Ping Wu; Christian Wiesmann; Sachdev S. Sidhu

We report a comprehensive analysis of sequence features that allow for the production of autonomous human heavy chain variable (VH) domains that are stable and soluble in the absence of a light chain partner. Using combinatorial phage-displayed libraries and conventional biophysical methods, we analyzed the entire former light chain interface and the third complementarity determining region (CDR3). Unlike the monomeric variable domains of camelid heavy chain antibodies (VHH domains), in which autonomous behavior depends on interactions between the hydrophobic former light chain interface and CDR3, we find that the stability of many in vitro evolved VH domains is essentially independent of the CDR3 sequence and instead derives from mutations that increase the hydrophilicity of the former light chain interface by replacing exposed hydrophobic residues with structurally compatible hydrophilic substitutions. The engineered domains can be expressed recombinantly at high yield, are predominantly monomeric at high concentrations, unfold reversibly, and are even more thermostable than typical camelid VHH domains. Many of the stabilizing mutations are rare in natural VH and VHH domains and thus could not be predicted by studying natural sequences and structures. The results demonstrate that autonomous VH domains with structural properties beyond the scope of natural frameworks can be derived by using non-natural mutations, which differ from those found in camelid VHH domains. These findings should enable the development of libraries of synthetic VH domains with CDR3 diversities unconstrained by structural demands.

Collaboration


Dive into the Helga Raab's collaboration.

Researchain Logo
Decentralizing Knowledge