Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: European Journal of Pharmacology, Elsevier BV, Vol. 820 ( 2018-02), p. 206-216
    Type of Medium: Online Resource
    ISSN: 0014-2999
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2018
    detail.hit.zdb_id: 1483526-5
    SSG: 15,3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: The Journal of Immunology, The American Association of Immunologists, Vol. 195, No. 5 ( 2015-09-01), p. 2343-2352
    Abstract: Macrophage migration inhibitory factor (MIF), a proinflammatory cytokine and counterregulator of glucocorticoids, is a potential therapeutic target. MIF is markedly different from other cytokines because it is constitutively expressed, stored in the cytoplasm, and present in the circulation of healthy subjects. Thus, the concept of targeting MIF for therapeutic intervention is challenging because of the need to neutralize a ubiquitous protein. In this article, we report that MIF occurs in two redox-dependent conformational isoforms. We show that one of the two isoforms of MIF, that is, oxidized MIF (oxMIF), is specifically recognized by three mAbs directed against MIF. Surprisingly, oxMIF is selectively expressed in the plasma and on the cell surface of immune cells of patients with different inflammatory diseases. In patients with acute infections or chronic inflammation, oxMIF expression correlated with inflammatory flare-ups. In addition, anti-oxMIF mAbs alleviated disease severity in mouse models of acute and chronic enterocolitis and improved, in synergy with glucocorticoids, renal function in a rat model of crescentic glomerulonephritis. We conclude that oxMIF represents the disease-related isoform of MIF; oxMIF is therefore a new diagnostic marker for inflammation and a relevant target for anti-inflammatory therapy.
    Type of Medium: Online Resource
    ISSN: 0022-1767 , 1550-6606
    RVK:
    RVK:
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2015
    detail.hit.zdb_id: 1475085-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 22, No. 5 ( 2023-05-04), p. 555-569
    Abstract: High levels of macrophage migration inhibitory factor (MIF) in patients with cancer are associated with poor prognosis. Its redox-dependent conformational isoform, termed oxidized MIF (oxMIF), is a promising tumor target due to its selective occurrence in tumor lesions and at inflammatory sites. A first-generation anti-oxMIF mAb, imalumab, was investigated in clinical trials in patients with advanced solid tumors, where it was well tolerated and showed signs of efficacy. However, imalumab has a short half-life in humans, increased aggregation propensity, and an unfavorable pharmacokinetic profile. Here, we aimed to optimize imalumab by improving its physicochemical characteristics and boosting its effector functions. Point mutations introduced into the variable regions reduced hydrophobicity and the antibodies’ aggregation potential, and increased plasma half-life and tumor accumulation in vivo, while retaining affinity and specificity to oxMIF. The introduction of mutations into the Fc region known to increase antibody-dependent cellular cytotoxicity resulted in enhanced effector functions of the novel antibodies in vitro, whereas reduced cytokine release from human peripheral blood mononuclear cells in the absence of target antigen by the engineered anti-oxMIF mAb ON203 versus imalumab reveals a favorable in vitro safety profile. In vivo, ON203 mAb demonstrated superior efficacy over imalumab in both prophylactic and established prostate cancer (PC3) mouse xenograft models. In summary, our data highlight the potential of the second-generation anti-oxMIF mAb ON203 as a promising immunotherapy for patients with solid tumors, warranting clinical evaluation.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    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. 75, No. 15_Supplement ( 2015-08-01), p. 2475-2475
    Abstract: Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine known to exacerbate tumor growth. MIF expression correlates with tumor aggressiveness and metastatic potential. We discovered a novel, disease-related conformational isoform of MIF which we have designated “oxMIF” because it can be mimicked in vitro by mild oxidation of recombinant MIF. A new class of fully human monoclonal antibodies (mAbs) specifically targeting oxMIF demonstrated efficacy in vivo and inhibited signaling pathways associated with tumor proliferation and progression in vitro. By developing novel immunohistochemistry (IHC) methods we were able to differentially detect oxMIF and to determine biodistribution of human anti-oxMIF mAbs in human and animal-derived tissue. First, we observed by IHC that in human pancreatic ductal adenocarcinoma (PDAC) tissue, oxMIF is specifically expressed in tumor cells and in tumor stroma within pancreatic intraepithelial lesions. In contrast, we could not detect oxMIF in healthy control tissue, whereas MIF (sum of oxMIF and its reduced isoform) was abundantly expressed in both, tissue from PDAC patients and tissue from healthy subjects. Then, in a murine genetic model of PDAC, we assessed the biodistribution of anti-oxMIF mAbs: intravenously injected I131-labelled anti-oxMIF mAbs significantly accumulated in the pancreas (primary tumor site), as well as in liver and lung (sites of metastasis). Subsequently, we investigated the biodistribution of anti-oxMIF mAbs in more detail using a syngeneic mouse model of chronic lymphocytic leukemia (eμ-myc CLL model): IHC analysis of lymph nodes from mAb treated mice confirmed co-localization of oxMIF and anti-oxMIF mAbs in cancerous tissue. Finally, we applied our IHC methods to assess the tissue penetration of anti-oxMIF mAb in liver metastases from late stage colorectal cancer patients (Ph 1 clinical study; ClinicalTrials.gov identifier: NCT01765790): we demonstrated that the antibody was able to penetrate metastases and to accumulate in tumor cells and in stromal tissue during the course of treatment. Our previous and current studies clearly demonstrate that oxMIF is a disease-related isoform of MIF that can be detected in malignant tissue and that treatment with fully human anti-oxMIF mAbs enables neutralization of oxMIF in tumor tissue. We therefore suggest that oxMIF reflects a potent new therapeutic target in solid tumors with diagnostic and prognostic value. A phase 1 clinical study of a novel fully human anti-oxMIF mAb is currently ongoing in patients with solid malignancies (ClinicalTrials.gov identifier: NCT01765790). Citation Format: Alexander Schinagl, Michael Thiele, Patrice Douillard, Deyaa Adib, Xiaochun Liu, Salim Yazji, Friedrich Scheiflinger, Randolf Kerschbaumer. A novel class of fully human monoclonal anti-oxMIF antibodies penetrates metastases and accumulates in tumor tissue. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 2475. doi:10.1158/1538-7445.AM2015-2475
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    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: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 14, No. 12_Supplement_2 ( 2015-12-01), p. A153-A153
    Abstract: Background: Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine known to exacerbate tumor growth. MIF expression correlates with tumor aggressiveness and metastatic potential. We discovered a disease related conformational isoform of MIF, which we designated “oxMIF” because it can be mimicked in vitro by mild oxidation of recombinant MIF (Thiele et al., 2015, J. Immunol.). We found oxMIF expressed in different types of cancer tissue but not in healthy control tissues. A new class of fully human monoclonal antibodies specifically targeting oxMIF (Kerschbaumer et al., 2012, J. Biol. Chem) demonstrated efficacy in vivo and inhibited tumor signaling pathways associated with tumor proliferation and progression in vitro (Hussain et al., 2013, Mol. Cancer Ther.). Imalumab is Baxalta's lead candidate anti-oxMIF antibody which is currently tested in a phase I clinical trial (ClinicalTrials.gov identifier: NCT01765790). Methods: Biodistribution of anti-oxMIF antibodies was assessed (i) in a murine genetic model of pancreatic cancer and (ii) in biopsies from phase 1 late stage patients with metastatic colorectal cancer (mCRC) who did receive imalumab intravenously (28-days cycles; 4 dose schedules) once weekly. Pre- and on-therapy tumor biopsies of CRC liver metastases from these treated patients were analyzed for antibody tissue penetration using newly developed immunohistochemistry (IHC) methods; in parallel biopsies were also analyzed for phosphoproteins and cytokines/chemokines content by multiplex. Results: In a murine genetic model of pancreatic cancer, intravenously injected I131-labelled anti-oxMIF antibody accumulated in the pancreas (primary tumor site), as well as in liver and lung (sites of metastasis). Analysis of biopsies from phase 1 patients with late stage mCRC revealed similar results as imalumab was able (i) to penetrate metastases, (ii) to co-localize with oxMIF in tumor cells and in stromal tissue and (iii) to accumulate during the course of treatment. While accumulating in tumor tissue, the antibody was cleared from the plasma with a half-life (half plasma steady-state concentration) of approximately 7 days. In parallel, analysis of intratumoral phosphoproteins and cytokines showed that imalumab down regulates tumorigenic and pro-inflammatory signals (e.g. Akt, MEK, MAPK, Btk) and pro-inflammatory cytokines (e.g. TNF-α and TNF-β) while up-regulating anti-inflammatory cytokines like IL-1ra and IL-10. Conclusion: Imalumab shows single agent antitumor activity in heavily pretreated patients with mCRC. These observations are in line with in vitro and animal studies using anti-oxMIF antibodies (Hussain et al., 2013, Mol. Cancer Ther.) and literature data on MIF tumorigenic functions. Based on these data, a 10 mg/kg dose has been recommended for a phase 2a study of imalumab in combination with 5-FU/Leucovorin or Panitumumab versus standard of care treatment in subjects with mCRC. Patient recruitment for this study is currently ongoing (ClinicalTrials.gov Identifier: NCT02448810) Citation Format: Patrice Douillard, Michael Thiele, Alexander Schinagl, Niels Halama, Dirk Jaeger, Salim Yazji, Friedrich Scheiflinger, Randolf Kerschbaumer. Imalumab, a first-in-class anti-oxidized macrophage migration inhibitory factor (oxMIF) antibody penetrates tumor tissues and shows antitumor activity in patients. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2015 Nov 5-9; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(12 Suppl 2):Abstract nr A153.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: European Journal of Pharmacology, Elsevier BV, Vol. 956 ( 2023-10), p. 175997-
    Type of Medium: Online Resource
    ISSN: 0014-2999
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2023
    detail.hit.zdb_id: 1483526-5
    SSG: 15,3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: The Lancet, Elsevier BV, Vol. 402, No. 10404 ( 2023-09), p. 798-808
    Type of Medium: Online Resource
    ISSN: 0140-6736
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2023
    detail.hit.zdb_id: 2067452-1
    detail.hit.zdb_id: 3306-6
    detail.hit.zdb_id: 1476593-7
    SSG: 5,21
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Oncotarget, Impact Journals, LLC, Vol. 7, No. 45 ( 2016-11-08), p. 73486-73496
    Type of Medium: Online Resource
    ISSN: 1949-2553
    URL: Issue
    Language: English
    Publisher: Impact Journals, LLC
    Publication Date: 2016
    detail.hit.zdb_id: 2560162-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2023
    In:  Cancer Research Vol. 83, No. 7_Supplement ( 2023-04-04), p. 585-585
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 585-585
    Abstract: Background: Radioimmunotherapy (RIT) of patients with solid tumors poses a challenge due to the slow blood clearance and delayed tumor uptake of directly radiolabeled antibodies that can cause high radiation exposure to normal tissues. Pretargeting techniques utilizing bispecific antibodies (bsmAbs) overcome these limitations by administering the tumor-targeting antibody and the radioactive payload sequentially. First, the bsmAb is injected and allowed to accumulate in the tumor. Once & gt;95% bsmAbs have cleared from circulation, a small radioligand with high affinity to the bispecific antibody is administered. Due to its pharmacokinetic profile, the radioligand quickly binds to the tumor-trapped bsmAbs while excess radioligand is rapidly excreted by the kidneys. Thereby, high tumor/non-tumor ratios are achieved. Oxidized Macrophage Migration Inhibitory Factor (oxMIF) is the disease-related structural isoform of the pleiotropic cytokine MIF and is specifically present in tumor tissue. Here we show preclinical efficacy of an anti-oxMIF x anti-HSG (histamine-succinyl-glycyl) bsmAb designed for pretargeted RIT (PRIT). Methods: Infrared dye-labeled cON-05, an anti-oxMIF x anti-HSG bsmAb, was administered to Balb/c mice bearing subcutaneous CT26 cancer syngrafts to assess tumor uptake and retention by infrared imaging and its pharmacokinetics by ELISA. A PRIT regimen using cON-05 in conjunction with a Lu177-loaded di-HSG peptide was examined in Balb/c mice bearing subcutaneous (s.c.) CT26 colon cancer syngrafts and in Balb/c nude mice bearing s.c. CFPAC-1 pancreatic cancer xenografts. Results: Infrared dye-labeled cON-05 accumulated in the tumor and was retained for & gt;7 days. cON-05 was found to clear from circulation following a biexponential decay with only 1-2% of the injected dose remaining in plasma after 3-5 days post-injection. Significant tumor regression and survival benefit (100% cON-05 vs. 0% vehicle) were achieved in mice bearing CT26 colorectal tumors, if cON-05 (5 mg/kg) was administered 3 days prior to applying Lu177-di-HSG. In the same model, when changing the pretargeting interval to 5 days, significant tumor growth inhibition (TGI; 29% and 44% on day 9) and survival benefit (15 and & gt;21 days cON-05 vs. 9 days vehicle) could be demonstrated for 2.5 and 5 mg/kg cON-05. In a CFPAC-1 pancreatic cancer mouse xenograft model, 38% TGI on day 28 was achieved upon injection of 5 mg/kg cON-05 followed by Lu177-di-HSG 5 days later. Conclusion: cON-05 is a novel anti-oxMIF x anti-HSG bsmAb designed for PRIT. cON-05 shows good tumor penetration and retention and tumor growth inhibition in conjunction with Lu177-di-HSG peptide in colorectal and pancreatic cancer mouse models. PRIT with cON-05 might offer new options for patients with difficult-to-treat tumors (e.g., pancreatic, gastric, head & neck) Citation Format: Alexander Schinagl, Irina Mirkina, Alejandro Puchol Tarazona. Pretargeted radioimmunotherapy with a novel anti-oxMIF/HSG bispecific antibody and a Lu177-loaded HSG radioligand results in significant tumor regression in murine models of cancer [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 585.
    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 ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 4841-4841
    Abstract: Macrophage migration inhibitory factor (MIF) was found to be upregulated in many cancers and to act in para- and autocrine loops within the tumor microenvironment. MIF has pleiotropic effects and plays a role in tumor growth via several mechanisms. (i) MIF acts directly on tumor cells by activating signaling pathways that promote cell proliferation and cell survival. (ii) MIF facilitates invasion of the extracellular matrix and induces angiogenesis and tumor vascularization. (iii) As a proinflammatory cytokine, MIF is one of the mediators of tumor micro-inflammation. However, MIF occurs in two immunologically distinct, redox-dependent isoforms. In its reduced form (redMIF), MIF is abundantly expressed and is present even in healthy subjects. In contrast, oxidized MIF (oxMIF) is found in patients with cancer. Highly selective, fully human monoclonal antibodies that specifically target oxMIF can counter-regulate the biological functions of MIF, indicating that oxMIF is the disease-related isoform of MIF and a relevant drug target. Animal studies and in vitro studies revealed that novel human antibodies that selectively target oxMIF, inhibit proliferation by reducing phosphorylation of ERK1/2 and AKT, promote apoptosis by activating caspase 3, inhibit angiogenesis and metastasis by reducing VEGF expression and blood vessel density within the tumor, and decrease cancer-associated inflammation of the tumor by downregulating the production of proinflammatory cytokines. We used MIF wild type (wt) and MIF knockout (KO) murine pancreatic cancer cells and MIF wt and MIF KO mice with C57Bl/6 background to dissect the contribution of stromal versus tumor derived MIF to tumor progression. Transfer of a pancreatic cancer cell line expressing MIF into wt mice or MIF KO mice resulted in aggressive tumor growth, liver metastasis, and survival of approximately 30 days in both models. Similar results were obtained by transferring a MIF KO pancreatic cancer cell line into MIF wt mice. Only transfer of a MIF KO cell line into MIF KO mice led to improved overall survival of approximately 80 days and completely abrogated metastasis. In a tumor xenograft model, an intravenously applied oxMIF-specific fully human antibody was able to penetrate the cancerous tissue and was detected in the stroma and the tumor. We conclude that both tumor stroma and tumor cells are an efficient source of oxMIF to promote tumor growth and metastasis, and that anti-oxMIF antibodies are able to neutralize oxMIF in the stroma and the tumor. A phase 1 clinical study of a novel human antibody that selectively targets oxMIF is currently ongoing in patients with solid malignancies (ClinicalTrials.gov identifier: NCT01765790). Citation Format: Alexander Schinagl, Thorsten Hagemann, Patrice Douillard, Michael Thiele, Dirk Voelkel, Michael Freissmuth, Friedrich Scheiflinger, Randolf J. Kerschbaumer. Oxidized macrophage migration inhibitory factor (oxMIF) expressed by tumor stroma and tumor cells, contributes to tumor growth. [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 4841. doi:10.1158/1538-7445.AM2014-4841
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages