Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    Online Resource
    Online Resource
    Proceedings of the National Academy of Sciences ; 2020
    In:  Proceedings of the National Academy of Sciences Vol. 117, No. 46 ( 2020-11-17), p. 28971-28979
    In: Proceedings of the National Academy of Sciences, Proceedings of the National Academy of Sciences, Vol. 117, No. 46 ( 2020-11-17), p. 28971-28979
    Abstract: Blocking the action of FSH genetically or pharmacologically in mice reduces body fat, lowers serum cholesterol, and increases bone mass, making an anti-FSH agent a potential therapeutic for three global epidemics: obesity, osteoporosis, and hypercholesterolemia. Here, we report the generation, structure, and function of a first-in-class, fully humanized, epitope-specific FSH blocking antibody with a K D of 7 nM. Protein thermal shift, molecular dynamics, and fine mapping of the FSH–FSH receptor interface confirm stable binding of the Fab domain to two of five receptor-interacting residues of the FSHβ subunit, which is sufficient to block its interaction with the FSH receptor. In doing so, the humanized antibody profoundly inhibited FSH action in cell-based assays, a prelude to further preclinical and clinical testing.
    Type of Medium: Online Resource
    ISSN: 0027-8424 , 1091-6490
    RVK:
    RVK:
    Language: English
    Publisher: Proceedings of the National Academy of Sciences
    Publication Date: 2020
    detail.hit.zdb_id: 209104-5
    detail.hit.zdb_id: 1461794-8
    SSG: 11
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: eLife, eLife Sciences Publications, Ltd, Vol. 11 ( 2022-09-20)
    Abstract: Pharmacological and genetic studies over the past decade have established the follicle-stimulating hormone (FSH) as an actionable target for diseases affecting millions, namely osteoporosis, obesity, and Alzheimer’s disease. Blocking FSH action prevents bone loss, fat gain, and neurodegeneration in mice. We recently developed a first-in-class, humanized, epitope-specific FSH-blocking antibody, MS-Hu6, with a K D of 7.52 nM. Using a Good Laboratory Practice (GLP)-compliant platform, we now report the efficacy of MS-Hu6 in preventing and treating osteoporosis in mice and parameters of acute safety in monkeys. Biodistribution studies using 89 Zr-labeled, biotinylated or unconjugated MS-Hu6 in mice and monkeys showed localization to bone and bone marrow. The MS-Hu6 displayed a β phase t ½ of 7.5 days (180 hr) in humanized Tg32 mice. We tested 217 variations of excipients using the protein thermal shift assay to generate a final formulation that rendered MS-Hu6 stable in solution upon freeze-thaw and at different temperatures, with minimal aggregation, and without self-, cross-, or hydrophobic interactions or appreciable binding to relevant human antigens. The MS-Hu6 showed the same level of “humanness” as human IgG1 in silico and was non-immunogenic in ELISpot assays for IL-2 and IFN-γ in human peripheral blood mononuclear cell cultures. We conclude that MS-Hu6 is efficacious, durable, and manufacturable, and is therefore poised for future human testing.
    Type of Medium: Online Resource
    ISSN: 2050-084X
    Language: English
    Publisher: eLife Sciences Publications, Ltd
    Publication Date: 2022
    detail.hit.zdb_id: 2687154-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Journal of the Endocrine Society, The Endocrine Society, Vol. 7, No. Supplement_1 ( 2023-10-05)
    Abstract: Disclosure: A.R. Pallapati: None. J. Gimenez-Roig: None. S. Rojekar: None. F. Korkmaz: None. D. Sant: None. O. Barak: None. F. Sultana: None. O. Moldavski: None. A. Gumerova: None. H.S. Kannangara: None. U. Cheliadinova: None. C.J. Rosen: None. J.N. Caminis: None. M. Meseck: None. V.E. Demambro: None. S.L. Sims: None. S. Gera: None. R. Witztum: None. S. Miyashita: None. V. Ryu: None. M. Saxena: None. T. Frolinger: None. A. Macdonald: None. S. Kim: None. G. Pevnev: None. D. Lizneva: None. T. Yuen: None. M. Zaidi: None. Pharmacological and genetic studies suggest that FSH is an actionable target for diseases affecting millions, notably osteoporosis, obesity and Alzheimer’s disease (AD). Blocking FSH action prevents bone loss (1, 2), fat accrual (3) and AD-like features in mice (4). We recently developed a first-in-class, humanized, epitope-specific FSH blocking antibody that binds to a 13-amino-acid-long sequence of FSHβ—MS-Hu6—with a KD of 7.52 nM (5). We showed that MS-Hu6 binds specifically to FSHβ, without binding to LH and TSH. For efficacy studies, we have blocked FSH action using either MS-Hu6 or the parent murine antibody, Hf2, targeted to the same epitope. Using our Good Laboratory Practice platform (Code of Federal Regulations, Title 21, Part 58), we report that FSH blockade prevents obesity, osteoporosis and AD in mice. We injected 20-week-old C57BL/6 male mice on a high-fat diet with a range of doses of Hf2 or vehicle s.c. five-days-a-week for 8 weeks. Hf2 (100 µg/mouse/day) reduced the increase in fat mass by 33% starting week 3, with a 7% reduction in in body weight. In separate studies, MS-Hu6 not only caused beiging of white adipose tissue in UCP1-reporter ThermoMice (IVIS imaging), but also improved bone density and microstructure (micro-CT) by elevating bone formation (dynamic histomorphometry). The increase in bone mass and improved microstructure were replicated in Cliff Rosen’s lab using C57BL6 mice 24 weeks post-ovariectomy. Novel Object Recognition testing of AD-prone, ovariectomized 3xTg mice showed a deficit in recognition memory, which was reversed after 8 weeks of Hf2 (100 µg/mouse/day for 5-days-a-week) exposure. Biodistribution studies using 89Zr-labelled, biotinylated or unconjugated MS-Hu6 in mice and monkeys showed localization to bone, bone marrow, fat depots and brain tissue. MS-Hu6 displayed a β phase t½ of 7.5 days in humanized Tg32 mice. In monkeys, an acute single injection of MS-Hu6 did not affect vitals, and biochemical parameters remained within the normative range. We tested 215 variations of excipients using a range of physicochemical techniques, including protein thermal shift, size exclusion chromatography, dynamic light scattering, Fourier-transform infrared spectroscopy, circular dichroism spectroscopy, and differential scanning calorimetry, to yield a formulation with thermal, colloidal, monomeric and structural stability at an ultra-high concentration (100 mg/mL) with acceptable viscosity, clarity and turbidity parameters. MS-Hu6 showed the same “humanness” as human IgG1 in silico and was non-immunogenic in ELISPOT assays for IL-2 and IFNγ in human PBMC cultures. In conclusion, MS-Hu6 is efficacious, durable and manufacturable, and is therefore poised for human testing as a multipurpose therapeutic for obesity, osteoporosis, and perhaps for AD. 1Cell, 2006; 2PNAS, 2018,; 3Nature, 2017; 4Nature, 2022; 5. PNAS, 2020, eLife, 2022. Presentation: Saturday, June 17, 2023
    Type of Medium: Online Resource
    ISSN: 2472-1972
    Language: English
    Publisher: The Endocrine Society
    Publication Date: 2023
    detail.hit.zdb_id: 2881023-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Journal of the Endocrine Society, The Endocrine Society, Vol. 7, No. Supplement_1 ( 2023-10-05)
    Abstract: Disclosure: S. Rojekar: None. A.R. Pallapati: None. J. Gimenez–Roig: None. D. Sant: None. S. Gera: None. F. Korkmaz: None. F. Sultana: None. O. Barak: None. O. Moldavski: None. U. Cheliadinova: None. A. Gumerova: None. S. Miyashita: None. H.S. Kannangara: None. T. Frolinger: None. R. Witztum: None. A. Macdonald: None. P. Georgii: None. S.L. Sims: None. J.N. Caminis: None. M. Meseck: None. V. Ryu: None. S. Kim: None. D. Lizneva: None. T. Yuen: None. M. Zaidi: None. Highly concentrated antibody formulations are oftentimes required for subcutaneous, self-administered biologics. Here, we report the creation of a unique formulation for our first-in-class FSH-blocking humanized antibody, MS-Hu6, which we propose to move to the clinic for osteoporosis, obesity, and Alzheimer’s disease. The studies were carried out using our Good Laboratory Practice (GLP) platform, compliant with the Code of Federal Regulations (Title 21, Part 58). We first used protein thermal shift, size exclusion chromatography, and dynamic light scattering to examine MS-Hu6 concentrations between 1 and 100 mg/mL. We found that thermal, monomeric, and colloidal stability of formulated MS-Hu6 was maintained at a concentration of 100 mg/mL. The addition of the antioxidant L-methionine and chelating agent disodium EDTA improved the formulation's long-term colloidal and thermal stability. Thermal stability was further confirmed by Nano differential scanning calorimetry (DSC). Physiochemical properties of formulated MS-Hu6, including viscosity, turbidity, and clarity, conformed with acceptable industry standards. That the structural integrity of MS-Hu6 in formulation was maintained was proven through Circular Dichroism (CD) and Fourier Transform Infrared (FTIR) spectroscopy. Three rapid freeze-thaw cycles at −80°C/25°C or −80°C/37°C further revealed excellent thermal and colloidal stability. Furthermore, formulated MS-Hu6, particularly its Fab domain, displayed thermal and monomeric storage stability for more than 90 days at 4°C and 25°C. Finally, the unfolding temperature (Tm) for formulated MS-Hu6 increased by & gt;4.80°C upon binding to recombinant FSH, indicating highly specific ligand binding. Overall, we document the feasibility of developing a stable, manufacturable and transportable MS-Hu6 formulation at a ultra-high concentration at industry standards. The study should become a resource for developing biologic formulations in academic medical centers. Presentation: Saturday, June 17, 2023
    Type of Medium: Online Resource
    ISSN: 2472-1972
    Language: English
    Publisher: The Endocrine Society
    Publication Date: 2023
    detail.hit.zdb_id: 2881023-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Annals of the New York Academy of Sciences, Wiley, Vol. 1521, No. 1 ( 2023-03), p. 67-78
    Abstract: Biopharmaceutical products are formulated using several Food and Drug Administration (FDA) approved excipients within the inactive ingredient limit to maintain their storage stability and shelf life. Here, we have screened and optimized different sets of excipient combinations to yield a thermally stable formulation for the humanized follicle‐stimulating hormone (FSH)–blocking antibody, MS‐Hu6. We used a protein thermal shift assay in which rising temperatures resulted in the maximal unfolding of the protein at the melting temperature ( T m ). To determine the buffer and pH for a stable solution, four different buffers with a pH range from 3 to 8 were screened. This resulted in maximal T m s at pH 5.62 for Fab in phosphate buffer and at pH 6.85 for Fc in histidine buffer. Upon testing a range of salt concentrations, MS‐Hu6 was found to be more stable at lower concentrations, likely due to reduced hydrophobic effects. Molecular dynamics simulations revealed a higher root‐mean‐square deviation with 1 mM than with 100 mM salt, indicating enhanced stability, as noted experimentally. Among the stabilizers tested, Tween 20 was found to yield the highest T m and reversed the salt effect. Among several polyols/sugars, trehalose and sucrose were found to produce higher thermal stabilities. Finally, binding of recombinant human FSH to MS‐Hu6 in a final formulation (20 mM phosphate buffer, 1 mM NaCl, 0.001% w/v Tween 20, and 260 mM trehalose) resulted in a thermal shift (increase in T m ) for the Fab, but expectedly not in the Fc domain. Given that we used a low dose of MS‐Hu6 (1 μM), the next challenge would be to determine whether 100‐fold higher, industry‐standard concentrations are equally stable.
    Type of Medium: Online Resource
    ISSN: 0077-8923 , 1749-6632
    URL: Issue
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2023
    detail.hit.zdb_id: 2834079-6
    detail.hit.zdb_id: 211003-9
    detail.hit.zdb_id: 2071584-5
    SSG: 11
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Journal of the Endocrine Society, The Endocrine Society, Vol. 6, No. Supplement_1 ( 2022-11-01), p. A883-A884
    Abstract: Pharmacological and genetic studies over the past decade suggest that FSH is an actionable target for diseases affecting millions, notably osteoporosis, obesity and Alzheimer's disease (AD). Blocking FSH action prevents bone loss (1, 2), fat and energy metabolism (3) and AD–like features in mice (4). We recently developed a first–in–class, humanized, epitope–specific FSH blocking antibody that binds to a 13–amino–acid–long sequence of FSHβ—"MS-Hu6"—with a KD of 7.52 nM (5). We showed that MS-Hu6 bound specifically to FSHβ and its different glycosylated forms, namely FSHβ21/18 and FSHβ24, without binding to LH and TSH. Here, using a GLP–compliant platform, we report the efficacy of MS-Hu6 in preventing obesity, osteoporosis and AD in mice. Notably, MS-Hu6-treated mice showed lower body weight and fat mass, increased lean mass (qNMR) and evidence of beiging in ThermoMice (IVIS imaging) compared with IgG–treated mice. Consistent with this, the thermogenic genes Ucp1 and Cidea were upregulated, whereas Pparg expression was attenuated in fat depots. Treatment of ThermoMice for 8 weeks also increased bone mineral density (BMD), improved microstructure (micro-CT), elevated bone formation (dynamic histomorphometry), and upregulated the osteoblastic genes Alp and Col1a1. The increase in bone mass and improved microstructure were replicated in C.J.R's lab using female mice 24 weeks post–ovariectomy. Preliminary testing using AD mice, namely APP/PS1 mice, showed that MS-Hu6 prevented the impairment in recognition and contextual memory. Biodistribution studies using 89Zr–labelled, biotinylated or unconjugated MS-Hu6 in mice and monkeys showed localization to bone, bone marrow and fat depots. MS-Hu6 displayed a β phase t½ of 13 days (316 hours) in humanized Tg32 mice, and bound endogenous FSH. In monkeys, an acute single injection of MS-Hu6 did not affect vitals, and biochemical parameters remained within the normative range. We tested 215 variations of excipients using the protein thermal shift assay to generate a final formulation that rendered MS-Hu6 stable in solution upon freeze–thaw and at different temperatures, with minimal aggregation, and without self–, cross–, or hydrophobic interactions or appreciable binding to relevant human antigens. MS-Hu6 showed the same level of "humanness" as human IgG1 in silico, and was non–immunogenic in ELISPOT assays for IL-2 and IFNγ in human peripheral blood mononuclear cell cultures. In conclusion, MS-Hu6 is efficacious, durable and manufacturable, and is therefore poised for future human testing as a multipurpose therapeutic for obesity, osteoporosis, and perhaps for AD.References: 1Sun et al., Cell, 2006, PMID: 16630814; Ji et al, PNAS, 2018, PMID: 29440419; 3Liu et al., Nature, 2017, PMID: 28538730; 4Xiong et al., Nature (In press); 5Gera et al., PNAS, 2020, PMID: 33127753 Presentation: Monday, June 13, 2022 12:30 p.m. - 2:30 p.m.
    Type of Medium: Online Resource
    ISSN: 2472-1972
    Language: English
    Publisher: The Endocrine Society
    Publication Date: 2022
    detail.hit.zdb_id: 2881023-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: eLife, eLife Sciences Publications, Ltd, Vol. 12 ( 2023-06-19)
    Abstract: Highly concentrated antibody formulations are oftentimes required for subcutaneous, self-administered biologics. Here, we report the development of a unique formulation for our first-in-class FSH-blocking humanized antibody, MS-Hu6, which we propose to move to the clinic for osteoporosis, obesity, and Alzheimer’s disease. The studies were carried out using our Good Laboratory Practice (GLP) platform, compliant with the Code of Federal Regulations (Title 21, Part 58). We first used protein thermal shift, size exclusion chromatography, and dynamic light scattering to examine MS-Hu6 concentrations between 1 and 100 mg/mL. We found that thermal, monomeric, and colloidal stability of formulated MS-Hu6 was maintained at a concentration of 100 mg/mL. The addition of the antioxidant L-methionine and chelating agent disodium EDTA improved the formulation’s long-term colloidal and thermal stability. Thermal stability was further confirmed by Nano differential scanning calorimetry (DSC). Physiochemical properties of formulated MS-Hu6, including viscosity, turbidity, and clarity, confirmed with acceptable industry standards. That the structural integrity of MS-Hu6 in formulation was maintained was proven through Circular Dichroism (CD) and Fourier Transform Infrared (FTIR) Spectroscopy. Three rapid freeze–thaw cycles at –80 °C/25 °C or –80 °C/37 °C further revealed excellent thermal and colloidal stability. Furthermore, formulated MS-Hu6, particularly its Fab domain, displayed thermal and monomeric storage stability for more than 90 days at 4°C and 25°C. Finally, the unfolding temperature (T m ) for formulated MS-Hu6 increased by 〉 4.80 °C upon binding to recombinant FSH, indicating highly specific ligand binding. Overall, we document the feasibility of developing a stable, manufacturable and transportable MS-Hu6 formulation at a ultra-high concentration at industry standards. The study should become a resource for developing biologic formulations in academic medical centers.
    Type of Medium: Online Resource
    ISSN: 2050-084X
    Language: English
    Publisher: eLife Sciences Publications, Ltd
    Publication Date: 2023
    detail.hit.zdb_id: 2687154-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Nature, Springer Science and Business Media LLC, Vol. 603, No. 7901 ( 2022-03-17), p. 470-476
    Type of Medium: Online Resource
    ISSN: 0028-0836 , 1476-4687
    RVK:
    RVK:
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2022
    detail.hit.zdb_id: 120714-3
    detail.hit.zdb_id: 1413423-8
    SSG: 11
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    Proceedings of the National Academy of Sciences ; 2020
    In:  Proceedings of the National Academy of Sciences Vol. 117, No. 25 ( 2020-06-23), p. 14386-14394
    In: Proceedings of the National Academy of Sciences, Proceedings of the National Academy of Sciences, Vol. 117, No. 25 ( 2020-06-23), p. 14386-14394
    Abstract: We report that two widely-used drugs for erectile dysfunction, tadalafil and vardenafil, trigger bone gain in mice through a combination of anabolic and antiresorptive actions on the skeleton. Both drugs were found to enhance osteoblastic bone formation in vivo using a unique gene footprint and to inhibit osteoclast formation. The target enzyme, phosphodiesterase 5A (PDE5A), was found to be expressed in mouse and human bone as well as in specific brain regions, namely the locus coeruleus, raphe pallidus, and paraventricular nucleus of the hypothalamus. Localization of PDE5A in sympathetic neurons was confirmed by coimmunolabeling with dopamine β-hydroxylase, as well as by retrograde bone-brain tracing using a sympathetic nerve-specific pseudorabies virus, PRV152. Both drugs elicited an antianabolic sympathetic imprint in osteoblasts, but with net bone gain. Unlike in humans, in whom vardenafil is more potent than tadalafil, the relative potencies were reversed with respect to their osteoprotective actions in mice. Structural modeling revealed a higher binding energy of tadalafil to mouse PDE5A compared with vardenafil, due to steric clashes of vardenafil with a single methionine residue at position 806 in mouse PDE5A. Collectively, our findings suggest that a balance between peripheral and central actions of PDE5A inhibitors on bone formation together with their antiresorptive actions specify the osteoprotective action of PDE5A blockade.
    Type of Medium: Online Resource
    ISSN: 0027-8424 , 1091-6490
    RVK:
    RVK:
    Language: English
    Publisher: Proceedings of the National Academy of Sciences
    Publication Date: 2020
    detail.hit.zdb_id: 209104-5
    detail.hit.zdb_id: 1461794-8
    SSG: 11
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages