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
    Elsevier BV ; 1987
    In:  Tetrahedron Letters Vol. 28, No. 17 ( 1987-1), p. 1873-1876
    In: Tetrahedron Letters, Elsevier BV, Vol. 28, No. 17 ( 1987-1), p. 1873-1876
    Type of Medium: Online Resource
    ISSN: 0040-4039
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 1987
    detail.hit.zdb_id: 2007074-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    Elsevier BV ; 2009
    In:  Bioorganic & Medicinal Chemistry Letters Vol. 19, No. 22 ( 2009-11), p. 6317-6318
    In: Bioorganic & Medicinal Chemistry Letters, Elsevier BV, Vol. 19, No. 22 ( 2009-11), p. 6317-6318
    Type of Medium: Online Resource
    ISSN: 0960-894X
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2009
    detail.hit.zdb_id: 1501505-1
    SSG: 15,3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Bioconjugate Chemistry, American Chemical Society (ACS), Vol. 31, No. 8 ( 2020-08-19), p. 1893-1898
    Type of Medium: Online Resource
    ISSN: 1043-1802 , 1520-4812
    Language: English
    Publisher: American Chemical Society (ACS)
    Publication Date: 2020
    detail.hit.zdb_id: 1500067-9
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 6294-6294
    Abstract: Introduction: The chemokine receptor CXCR5 is highly expressed in tumor cells from different lymphoma types and represents a viable drug target for the development of antibody-drug conjugates (ADC) to treat patients with lymphoma.   Methods: Immunohistochemistry of human tissues of different lymphoma types were stained using antibodies against human CXCR5, CD20, CD70 and CD79b. Slides were examined by a pathologist and scored for expression. CXCR5 expression on tumor cell lines was analyzed by flow cytometry. Cell surface receptor density was analyzed by Quantibrite ™ PE-beads. Evaluation of antibody internalization was performed using the Operetta High Content Imaging System. NOD/SCID mice were transplanted subcutaneously with lymphoma patient-derived (PDX) tumor specimens and treated with 2mg/kg or 10mg/kg VIP924 CXCR5-ADC or isotype control. Results: CXCR5 reveals very low to no expression in most tissues except for lymph nodes. Expression of CXCR5 and other B-cell targets like CD19, CD20, CD70 and CD79b was analyzed on patient-derived tumor samples. High expression of CXCR5 was found on naïve and previously treated diffuse large B-cell lymphoma (DLBCL), follicular lymphoma (FL), mantle cell lymphoma (MCL), and chronic lymphocytic leukemia (CLL) samples. Based on these results, we developed a CXCR5 targeted ADC with a novel, highly potent kinesin spindle protein inhibitor (KSPi) payload linked via a legumain-cleavable linker (VIP924). To compare the performance of KSPi ADCs, the same effector chemistry was attached to antibodies against other B-cell targets and tested in cytotoxicity assays in various cell lines. CXCR5-targeted ADC demonstrates significantly higher activity compared to the other ADCs tested except for the CD79b ADC with equal potency. VIP924 was then tested in vivo in different PDX tumor mouse models with different levels of CXCR5 expression. In the LY2264 and HBL-1 DLBCL models, we obtained a tumor growth inhibition of 68% and 100% respectively compared to isotype control. The median survival time for the isotype control group was 29 days and median survival time for the mice treated with 10mg/kg VIP924 could not be determined as all mice survived until the end of the study (Day 37). No effect on body weight or any adverse effects in the VIP924 treated mice were observed. Conclusions: CXCR5 is a highly attractive target in hematological malignancies such as DLBCL, MCL, and FL due to high protein expression and almost no expression in healthy tissues. CXCR5-targeting ADC with KSPi payload showed high potency and superiority to other B-cell-targeted ADCs in vitro on a broad range of lymphoma cell lines. VIP924 with a novel legumain-cleavable linker showed activity in in vivo PDX models from lymphoma patients. Due to the high CXCR5 expression found in relapsed DLBCL patients, VIP924 may bring promising new treatment options for previously treated patients with lymphoma. Citation Format: Tibor Schomber, Beatrix Stelte-Ludwig, Amy J. Johnson, Oliver von Ahsen, Christoph Schatz, Raquel Izumi, Ahmed Hamdy, Hans-Georg Lerchen. CXCR5 is a very promising drug target for the development of antibody-drug conjugates to treat patients with lymphoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 6294.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: ChemMedChem, Wiley, Vol. 12, No. 10 ( 2017-05-22), p. 728-737
    Abstract: Adenosine is known to be released under a variety of physiological and pathophysiological conditions to facilitate the protection and regeneration of injured ischemic tissues. The activation of myocardial adenosine A 1 receptors (A 1 Rs) has been shown to inhibit myocardial pathologies associated with ischemia and reperfusion injury, suggesting several options for new cardiovascular therapies. When full A 1 R agonists are used, the desired protective and regenerative cardiovascular effects are usually overshadowed by unintended pharmacological effects such as induction of bradycardia, atrioventricular (AV) blocks, and sedation. These unwanted effects can be overcome by using partial A 1 R agonists. Starting from previously reported capadenoson we evaluated options to tailor A 1 R agonists to a specific partiality range, thereby optimizing the therapeutic window. This led to the identification of the potent and selective agonist neladenoson, which shows the desired partial response on the A 1 R, resulting in cardioprotection without sedative effects or cardiac AV blocks. To circumvent solubility and formulation issues for neladenoson, a prodrug approach was pursued. The dipeptide ester neladenoson bialanate hydrochloride showed significantly improved solubility and exposure after oral administration. Neladenoson bialanate hydrochloride is currently being evaluated in clinical trials for the treatment of heart failure.
    Type of Medium: Online Resource
    ISSN: 1860-7179 , 1860-7187
    URL: Issue
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2017
    detail.hit.zdb_id: 2209649-8
    SSG: 15,3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    Wiley ; 1991
    In:  Angewandte Chemie International Edition in English Vol. 30, No. 12 ( 1991-12), p. 1682-1684
    In: Angewandte Chemie International Edition in English, Wiley, Vol. 30, No. 12 ( 1991-12), p. 1682-1684
    Type of Medium: Online Resource
    ISSN: 0570-0833 , 1521-3773
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 1991
    detail.hit.zdb_id: 2011836-3
    detail.hit.zdb_id: 123227-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 5445-5445
    Abstract: C4.4a (LYPD3) has been identified previously as a cancer- and metastasis-associated internalizing cell surface protein. Targeting C4.4a with a specific antibody-drug conjugate (ADC) represents an unique opportunity to treat tumors with high unmet medical need such as squamous cell carcinomas SCC, in particular lung SCC. We have generated an anti-C4.4a ADC consisting of a fully human monoclonal antibody linked to a non cell-permeable tubulin-binding auristatin cytotoxic agent (technology licensed from Seattle Genetics). In vitro, anti-C4.4a ADC showed an anti-proliferative efficacy (IC50) in the nanomolar range in cell lines endogenously expressing C4.4a (e.g. human lung cancer cell lines NCI-H292 and NCI-H322). High ADC stability and selectivity was observed in transfected A549 lung cancer cells over-expressing C4.4a compared to mock-transfected cells. In vivo, anti-C4.4a ADC exhibited a potent and selective antitumor activity in various human xenograft models (NCI-H292, NCI-H322, SCC-4) as well as in two SCC (Lu7433, Lu7343) and one pleomorphic (Lu7064) patient-derived lung cancer xenograft models. The in vivo efficacy is strictly target-dependent and selective as no efficacy was observed in C4.4a negative models (Fadu, Lu 7700) or using a non-specific isotype antibody ADC (NCI-H292, NCI-H322). A minimal effective dose (MED) as low as 1.9 mg/kg, response rates of up to 100%, and additive anti-tumor efficacy in combination with cisplatin were observed in the NCI-H292 xenograft model. Furthermore, it has been demonstrated that NCI-H292 were still sensitive to ADC treatment when tumors were allowed to regrow after the initial treatment cycle(). The anti-C4.4a ADC, which is fully cross-reactive with the mouse orthologue of C4.4a, was well tolerated at efficacious doses. Reversible skin reddening was observed only at doses markedly higher than the MED. In summary, anti-C4.4a ADC is a promising therapeutic candidate for the treatment of C4.4a-expressing squamous cell carcinomas, andpreclinical development has been initiated. Citation Format: Joerg Willuda, Lars Linden, Hans-Georg Lerchen, Charlotte Kopitz, Sven Golfier, Ute Bach, Joachim Schumacher, Beatrix Stelte-Ludwig, Oliver Von Ahsen, Claudia Schneider, Frank Dittmer, Rudolf Beier, Sherif El-Sheik, Jan Tebbe, Gabriele Leder, Heiner Apeler, Rolf Jautelat, Bertolt Kreft, Karl Ziegelbauer. Preclinical anti-tumor efficacy of an anti-C4.4a (LYPD3) antibody drug conjugate for the treatment of lung squamous cell carcinoma. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 5445. doi:10.1158/1538-7445.AM2014-5445
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 21 ( 2016-11-01), p. 6331-6339
    Abstract: The fibroblast growth factor receptor FGFR2 is overexpressed in a variety of solid tumors, including breast, gastric, and ovarian tumors, where it offers a potential therapeutic target. In this study, we present evidence of the preclinical efficacy of BAY 1187982, a novel antibody–drug conjugate (ADC). It consists of a fully human FGFR2 monoclonal antibody (mAb BAY 1179470), which binds to the FGFR2 isoforms FGFR2-IIIb and FGFR2-IIIc, conjugated through a noncleavable linker to a novel derivative of the microtubule-disrupting cytotoxic drug auristatin (FGFR2-ADC). In FGFR2-expressing cancer cell lines, this FGFR2-ADC exhibited potency in the low nanomolar to subnanomolar range and was more than 100-fold selective against FGFR2-negative cell lines. High expression levels of FGFR2 in cells correlated with efficient internalization, efficacy, and cytotoxic effects in vitro. Pharmacokinetic analyses in mice bearing FGFR2-positive NCI-H716 tumors indicated that the toxophore metabolite of FGFR2-ADC was enriched more than 30-fold in tumors compared with healthy tissues. Efficacy studies demonstrated that FGFR2-ADC treatment leads to a significant tumor growth inhibition or tumor regression of cell line–based or patient-derived xenograft models of human gastric or breast cancer. Furthermore, FGFR2 amplification or mRNA overexpression predicted high efficacy in both of these types of in vivo model systems. Taken together, our results strongly support the clinical evaluation of BAY 1187982 in cancer patients and a phase I study (NCT02368951) has been initiated. Cancer Res; 76(21); 6331–9. ©2016 AACR.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 46-46
    Abstract: Antibody-drug conjugates (ADCs) are promising agents that are developed for targeted delivery of cytotoxic payloads to tumor cells. ADCs share a common design of antibody, linker, and cytotoxic payload. Despite significant efforts, the number of available payload classes with a differentiated mode-of-action that can successfully be employed to generate antibody-drug conjugates (ADCs) is still rather limited. So far, only ADCs with microtubule depolymerizing or DNA binding payloads have been approved. The identification of ADC payload classes with a novel mode-of-action will increase therapeutic options and potentially help to overcome resistance. Inhibitors of the kinesin spindle protein (KSP/Eg5/KIF11) have generated interest due to their high anti-tumor activity. However, the transfer of the potency of small molecule KSP inhibitors (KSPis) to highly efficient clinical regimens with a sufficient therapeutic window remains challenging. Through the conjugation of a novel pyrrole subclass of KSPis to antibodies targeting different cancer antigens, we generated a panel of ADCs and characterized them both in vitro and in vivo. ADCs targeting either EGFR or TWEAKR/Fn14 showed strong and specific internalization and displayed specific and potent anti-proliferative efficacy in vitro. In cytotoxicity assays, these ADCs exhibited sub-nanomolar potency in antigen-positive cancer cell lines (EGFR/TWEAKR-pos. NCI-H292; TWEAKR-pos. BxPC3, LoVo) and more than 100-fold selectivity versus non-targeted control-ADC containing the same linker and the same payload. Furthermore, selective anti-tumor efficacy of EGFR- and TWEAKR-KSPi-ADCs was demonstrated in vivo using both cancer cell line-derived models of NSCLC (NCI-H292), urothelial cell carcinoma (UCC) (KU-19-19), and renal cell carcinoma (A498), as well as in the TWEAKR-positive patient-derived xenograft UCC model BFX469. At doses of 5-10 mg/kg qw or bw potent anti-tumor efficacy with treated-to-control ratios (T/C) between 0.16 to 0.28 as well as complete regressions were observed. In summary, KSP inhibitors have been established as a promising new payload class allowing the generation of highly potent and selective ADCs for the treatment of solid tumors. Citation Format: Anette Sommer, Sandra Berndt, Hans-Georg Lerchen, Beatrix Stelte-Ludwig, Sven Wittrock, Anne-Sophie Rebstock, Lisa Dietz, Christoph Mahlert, Simone Greven, Nils Griebenow, Yolanda Cancho-Grande, Rolf Jautelat, Heiner Apeler, Bertolt Kreft. Preclinical activity of novel antibody-drug conjugates with pyrrole-based kinesin spindle protein inhibitors targeting different tumor antigens [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 46. doi:10.1158/1538-7445.AM2017-46
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 2722-2722
    Abstract: The development of small molecule drug conjugates (SMDCs) is gaining momentum after the 2018 approval of the small molecule radioconjugate lutetium-177 vipivotide tetraxetan. In contrast to the use of antibodies for tumor homing, small molecule binding ligands may result in a better tumor penetration. Also the potential targets for ADCs are limited to internalizing antigens so the ADC can be trafficked to the lysosomes for release and activation of the payload. We sought new mechanisms for extracellular activation of SMDCs using enzymes, which are present in the tumor microenvironment (TME). For example, neutrophil elastase (NE) belongs to a family of serine proteases that degrades elastin and other extracellular matrix proteins. NE also contributes to cancer progression by enhancing tumor evasion and metastasis. NE expression and neutrophil tumor infiltration have been correlated with metastatic potential and poor prognosis. Previous studies have shown that NE efficiently cleaves and activates camptothecin conjugates by cleaving an ester bond1. Here we describe the successful use of NE for efficient activation and traceless payload release from several unique and proprietary SMDCs. Binding molecules such as a peptidomimetic αvβ3 ligand were linked via appropriate spacers to substrate tripeptides of NE. The C-terminal end was directly attached to different functional groups at various payloads such as camptothecins (CPT), monomethyl auristatin E, kinesin spindle protein inhibitors, and CDK9 inhibitors. Hindered ester bonds, amide bonds, and sulfoximide bonds were found to be efficiently cleaved by NE; conversely, those linkages were highly stable in culture medium and in circulation. Consequently, respective SMDCs are not or are moderately cytotoxic when incubated alone with cancer cell lines. However, in the presence of NE, cytotoxicity of the SMDCs increases by two orders of magnitude to the low nanomolar or subnanomolar range across several cell lines reaching similar potency as the respective payloads alone. In PK studies in mouse, rat, and dog, the SMDC with a CPT payload, shows a half-life of 3.6, 8.3, and 4.2 h, respectively, and excellent stability in circulation over 24 hr with low exposure levels ( & lt;1% of SMDC) of cleaved payload detectable. In an MX1 triple-negative breast cancer xenograft model, tumor regressions were observed when the SMDC was administered in a 2 days on/5 days off schedule. NE is capable of cleaving sterically hindered ester bonds, amides, and sulfoximides to release a variety of potent payloads from different SMDCs in a traceless manner. These molecules represents a versatile strategy for selective delivery of payloads to TME that does not require the tumor target to internalize. 1Lerchen HG, Stelte-Ludwig B, Kopitz C, et al. Cancers 2022 Citation Format: Hans-Georg Lerchen, Anne-Sophie Rebstock, Marieke Wiedmann, Beatrix Stelte-Ludwig, Harvey Wong, Dmitry Zubov, Raquel Izumi, Ahmed Hamdy. Synthesis and characterization of novel small molecule drug conjugates with different payloads designed to be released in tumor microenvironment by neutrophil elastase [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 2722.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    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