GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 13_Supplement ( 2021-07-01), p. 387-387
    Abstract: Introduction: Protein glycosylation is the most abundant and most complex form of post-translational protein modification. Glycosylation profoundly affects protein structure, conformation, and function. The elucidation of the potential role of differential protein glycosylation as biomarkers has so far been limited by the technical complexity of generating and interpreting this information. We have recently established a novel, powerful platform that combines ultra-high-performance liquid chromatography coupled to triple quadrupole mass spectrometry with a proprietary machine learning and neural-network-based data processing engine that allows, for the first time, high-throughput, highly scalable interrogation of the glycoproteome. Experimental Procedures: Using this platform we interrogated 413 individual glycopeptide (GP) signatures derived from 69 abundant serum proteins in pretreatment blood samples from a cohort of 36 individuals (11 females, 25 males, age range 28 to 90 years) with metastatic malignant melanoma treated either with nivolumab plus ipilimumab (12 patients) or pembrolizumab (24 patients). Progression-free survival (PFS) data with follow-up of up to 3.7 years (median: 0.8 years) were used as clinical endpoint phenotype against which the predictive power of differential abundance of GPs was assessed. PFS data were analyzed using Cox Proportional Hazards models, and Kaplan Meier curves were generated for GP markers that showed statistically significant differential abundances using an FDR-adjusted p-value of ≤0.1 as a cutoff. Summary of Results: We identified 27 GPs with abundance differences at FDR p≤0.1, and among them 8 at p≤0.001. Using the latter 8 markers, we created a multivariable model for PFS by generating leave-one-out cross-validation (LOOCV) scores and determining an optimized cutoff value for these scores using Harrel's concordance index. Dichotomizing the LOOCV scores using this cutoff value demonstrated the model to yield a hazard ratio of 9.2 at a p-value of 10-5 for separating treatment responders and non-responders (70% vs. 0% PFS, respectively, at 18 months based on LOOCV score above/below cutoff), as compared to a hazard ratio of 1.5, p=0.5 for PDL1 expression. Conclusions: Our results indicate that glycoproteomics holds a strong promise as a response predictor to checkpoint inhibitor treatment that appears to significantly outperform other currently pursued biomarker approaches in this context. Citation Format: Gege Xu, Rachel Rice, Hector Huang, Klaus Lindpaintner, Jillian M. Prendergast, Karl Normington, Dennie Frederick, Genevieve M. Boland, Daniel Serie. Glycoproteomics as a powerful liquid biopsy-based predictor of checkpoint-inhibitor treatment response [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 387.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 24, No. 15_Supplement ( 2018-08-01), p. B28-B28
    Abstract: Ovarian cancer is the leading cause of death from gynecologic malignancies in the U.S. The current standard of care is tumor debulking followed by chemotherapy. This treatment results in approximately 70% of patients achieving an initial complete clinical response. However, many of these patients will unfortunately relapse with chemoresistant disease developing in part to the presence of cancer stem cells (CSCs) within the tumor. Indeed, ovarian CSCs have been identified and shown to be resistant to chemo- and radiotherapy. Cancer cell surface glycans, called tumor-associated carbohydrate antigens (TACAs), are a class of attractive cancer-specific targets found on the cell surface of many solid tumors. Siamab is targeting a glycan structure (Sialyl-Tn, STn) that is cancer-specific and a major reported constituent of two well-known CSC biomarkers, CD44 and MUC1, residing on both CSCs and mature malignant cells in some cancer types. Although CD44 and MUC1 are also present on normal tissue and normal stem cell surfaces, tumor and CSC specificity is conferred the presence of STn-glycosylated variants of these CSC markers and other cell surface proteins that are essentially absent from normal human tissues. Importantly, STn can be a component of CA-125 (MUC16), conferring additional specificity to this diagnostic marker for ovarian carcinoma. The elevated presence of STn in tumors is associated with metastatic disease, poor prognosis, and reduced overall survival. In addition, elevations in STn expression are linked to chemotherapy resistance and enable tumors to evade the host immune system. We have observed that STn levels are elevated in cancer cells following chemotherapy treatment in both in vitro and in vivo model systems and STn is present on specific subtype of tumor-infiltrating myeloid-derived suppressor cells. Both observations support the role of STn in tumor chemoresistance and immunomodulation. While attempts have been made to target STn clinically with a cancer vaccine, efficacy has been limited. Siamab has developed a unique immunotherapeutic solution aimed at eradicating human neoplasias by targeting both CSCs and bulk tumor to enable whole tumor killing. We have identified and humanized a panel of mouse monoclonal novel antibodies (Abs) that specifically target with high affinity the STn glycan independent of its carrier protein. These Abs are highly selective in binding assays demonstrating robust nanomolar EC50s, possess specificity on tissues as determined with neuraminidase treatment, and exhibit STn-specific glycan binding on Siamab’s proprietary glycan array. Antitumor efficacy was evaluated in vitro and in vivo utilizing humanized anti-STn antibody drug conjugated (ADC) material. Cell cytotoxicity was demonstrated in a panel of STn-expressing cell lines with low nanomolar IC50s. We confirmed that the decrease in proliferation in vitro can be translated to a reduction in tumor size in vivo, through a series of cell line and patient-derived ovarian cancer xenograft models. Inhibition of tumor progression were observed in all models, with complete regressions observed in some treatment arms. No significant weight loss was observed for any treatment groups, indicating the therapy was well tolerated by all the groups. A decrease in STn expression was noted in the tumors following treatment, indicating that we were hitting the target against which these Abs were generated. In addition, we have begun the development of both tissue- and serum-based biomarker assays utilizing these selective anti-STn Abs. Our data demonstrate that high-affinity, STn-selective humanized mAbs show promise as therapies for ovarian tumors, as well as tools for patient stratification and pharmacodynamic biomarker assessments. Citation Format: Daniel T. Dransfield, Jillian M. Prendergast, David A. Eavarone, Rawan Nazer, Linah Al-Alem, Jenna Stein, Jeff Behrens, Bo Rueda. Targeting the tumor-associated carbohydrate antigen STn with humanized anti-Sialyl-Tn monoclonal antibody-drug conjugates inhibits ovarian cancer tumor growth in vitro and in vivo. [abstract]. In: Proceedings of the AACR Conference: Addressing Critical Questions in Ovarian Cancer Research and Treatment; Oct 1-4, 2017; Pittsburgh, PA. Philadelphia (PA): AACR; Clin Cancer Res 2018;24(15_Suppl):Abstract nr B28.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 36-36
    Abstract: Tumor-associated carbohydrate antigens (TACAs) historically have been challenging targets for antibody therapeutics. Sialyl-Tn (STn) is a cancer specific antigen that is expressed on the cell surface of carcinomas including ovarian, colon, prostate, and pancreatic tumors but is rarely present in normal tissue. STn expression has been linked to innate immune suppression, a chemoresistant phenotype, metastasis, and poor prognosis. Previous attempts to target this antigen in the clinic with synthetic glycan vaccines proved safe but lacked efficacy. We have developed highly selective humanized monoclonal antibodies and antibody drug conjugates (ADCs) targeting TACAs, such as STn. Remarkable sequence homology across all anti-STn mAbs was observed in both heavy and light chains, and hot spots for hypermutation were identified. These antibodies were selected using our glycan microarray that enriches for candidates whose binding is protein-independent, highly selective and demonstrates exceptional target affinity. Lead humanized candidates demonstrated single digit nanomolar EC50s in ELISA/flow cytometric assays, STn selective cell internalization, and STn specific glycan binding on Siamab’s proprietary glycan array. STn binding sites in common tumor lines (ovarian, gastric and breast) were determined per cell and subsequent cytotoxicity assays in these lines demonstrated in vitro efficacy. Tumor microarray experiments revealed membranous staining in cancerous tissues of various indications. Binding studies of anti-STn antibodies to primary human cancer samples by flow cytometry demonstrated that both tumor and Myeloid-Derived Suppressor Cells (MDSC, both myeloid and granulocytic) express STn. In an OVCAR3 xenograft model, 30 days after the last anti-STn ADC dose was given, groups treated (Q7Dx4) exhibited mean tumor volumes below the Day 1 pre-treatment mean tumor volumes (155mm3). Flow cytometric analysis of tumors from these mice demonstrated that anti-STn ADC treatment reduces STn expression on the primary tumor in a dose-dependent manner (Q7Dx4 vs. single dose) compared to the isotype-ADC control. Our data demonstrates that high-affinity, STn-selective mAbs show promise as therapies for solid tumors and could also target MDSCs to promote antitumor immune responses. Citation Format: Jillian M. Prendergast, David A. Eavarone, Patricia E. Rao, Adam D. Curtis, Lindsay S. Shopland, Todd A. Hoffert, Jenna Stein, Jeff Behrens, Daniel T. Dransfield. Novel anti-Sialyl-Tn monoclonal antibodies and antibody-drug conjugates (ADCs) demonstrate tumor specificity in vitro and in vivo antitumor efficacy [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 36. doi:10.1158/1538-7445.AM2017-36
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 5625-5625
    Abstract: Introduction: Myeloid derived suppressor cells (MDSCs) are functionally defined by their capacity to suppress T cell immunity; therefore inhibiting these cells is of great interest for immuno-oncology applications. STn, the sialylated version of the carbohydrate Tn antigen, is broadly expressed in human cancers such as breast, ovarian, bladder, cervical, colon, and lung and is rarely expressed in normal adult human tissues. The presence of STn in tumors is associated with metastatic disease, poor prognosis, and reduced overall survival. Tumor STn expression is well established and can be leveraged for targeted cancer therapy, however its expression on immuno-suppressive tumor infiltrating lymphocytes such as MDSCs has not been established. Methods: We generated humanized anti-STn mAbs having no cross-reactivity to the asialylated form of STn (Tn) or other glycan antigens. The epitope targeted by these mAbs is the STn glycan itself, not a particular glycopeptide or carrier protein, which offers the broadest potential to bind to multiple STn-glycosylated proteins on cell surfaces. We used these mAbs to quantify specifically the expression of tumor and MDSC STn expression across a set of patient tumor samples. We further treated mice with a humanized anti-STn-MMAE antibody-drug conjugate to evaluate targeted depletion of STn+ MDSCs. In order to evaluate the therapeutic potential of targeting this MDSC phenotype, we further evaluated the immunosuppressive environment of tumors containing STn+ and STn- MDSCs. Results: For the first time, we report the detection of STn on the surface of MDSCs in a growing body of patient tumor samples. Our data in patients as well as xenograft models additionally links STn expression on MDSCs to tumor cell STn expression. We have also demonstrated depletion of STn+ MDSCs after treatment with an anti-STn antibody-drug conjugate in an ovarian carcinoma xenograft model. Conclusions: Our data demonstrated that STn provides a uniquely glycan-specific and potent target for treatment of solid tumors. Anti-STn therapeutics offer the potential to go beyond tumor targeting to also directly target and deplete immune-suppressive MDSCs, thus fostering immune re-engagement and increasing the potential for better patient outcomes. Citation Format: David A. Eavarone, Patricia E. Rao, Jillian M. Prendergast, Adam D. Curtis, Rong Zhang, Lindsay S. Shopland, Jenna N. Stein, Jeff Behrens, Daniel T. Dransfield. Myeloid derived suppressor cells (MDSCs) express Sialyl Tn (STn) and are a therapeutic target for anti-STn antibody drug conjugates [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 5625.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: mAbs, Informa UK Limited, Vol. 9, No. 4 ( 2017-05-19), p. 615-627
    Type of Medium: Online Resource
    ISSN: 1942-0862 , 1942-0870
    Language: English
    Publisher: Informa UK Limited
    Publication Date: 2017
    detail.hit.zdb_id: 2537838-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease, Elsevier BV, Vol. 1870, No. 7 ( 2024-10), p. 167315-
    Type of Medium: Online Resource
    ISSN: 0925-4439
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2024
    detail.hit.zdb_id: 2209528-7
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2018
    In:  Molecular Cancer Therapeutics Vol. 17, No. 1_Supplement ( 2018-01-01), p. B114-B114
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 17, No. 1_Supplement ( 2018-01-01), p. B114-B114
    Abstract: Targeted therapeutics that can differentiate between normal and malignant tumor cells represent the ideal standard for the development of a successful anticancer strategy. The Sialyl-Thomsen-nouveau antigen (STn or Sialyl-Tn, also known as CD175s) is rarely observed in normal adult tissues, but it is abundantly expressed in many types of human epithelial cancers. Interestingly, we have observed that the level of this carbohydrate is elevated following chemotherapy treatment in both in vitro and in vivo model systems, suggesting a potential role of STn in chemoresistance. We have identified and humanized a panel of mouse monoclonal novel antibodies (Abs) that specifically target with high affinity the STn glycan independent of its carrier protein. We have demonstrated that these Abs are highly selective in binding assays demonstrating robust nanomolar EC50s, possess specificity on cancer tissues as determined with neuraminidase treatment, and exhibit STn-specific glycan binding on Siamab’s proprietary glycan array. Antitumor efficacy was evaluated in vitro and in vivo utilizing humanized anti-STn antibody-drug conjugated (ADC) material using MMAE as the cytotoxic agent. Cell cytotoxicity was demonstrated in a panel of STn-expressing cell lines with low nanomolar IC50s. In vivo, single-dose mouse PK studies were performed with humanized anti-STn ADCs and we determined half-life of approximately 3 days for both SIA01-ADC and SIA02-ADC. We confirmed that the decrease in proliferation in vitro can be translated to a reduction in tumor size in vivo through a series of cell line-derived and patient-derived (PDX) ovarian cancer and pancreatic cancer xenograft models. Inhibition of tumor progression was observed in all models with complete regressions observed in some treatment arms. No significant weight loss occurred in any treatment groups, indicating the therapy was well tolerated by all the groups. A decrease in STn expression was noted in the tumors following treatment, indicating that we were hitting the target against which these Abs were generated. In addition to these preclinical pharmacology studies, we demonstrated that the administration of our anti-STn ADC has an excellent safety profile through the completion of a non-GLP pilot PK/toxicity study in cynomolgus monkeys. No weight loss or deaths occurred in this study and no gross pathology changes were observed in all organs examined. Histopathologic changes were limited to the bone marrow with minimal to mild decreased cellularity and mild decrease in the myeloid to erythroid ratio observed. All clinical chemistry results (liver, kidney function, etc.) were normal throughout study. Changes in hematology parameters–modest neutropenia–were consistent with other MMAE ADCs and therefore, we conclude, are not related to the STn target, which is consistent with our tissue cross-reactivity studies demonstrating that overall normal tissue expression of the STn target is insignificant. Our data demonstrate that high-affinity, STn-selective humanized mAbs show promise as therapies for solid tumors. Citation Format: Daniel Dransfield, Jillian M. Prendergast, David A. Eavarone, Rawan Nazer, Linah Al-Alem, Bo Rueda, Jenna Stein, Jeff Behrens. Humanized anti-Sialyl-Tn monoclonal antibody-drug conjugates inhibit tumor growth in vitro and in vivo [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2017 Oct 26-30; Philadelphia, PA. Philadelphia (PA): AACR; Mol Cancer Ther 2018;17(1 Suppl):Abstract nr B114.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Immunology, Immunotherapy, Springer Science and Business Media LLC, Vol. 67, No. 9 ( 2018-9), p. 1437-1447
    Type of Medium: Online Resource
    ISSN: 0340-7004 , 1432-0851
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2018
    detail.hit.zdb_id: 1458489-X
    detail.hit.zdb_id: 195342-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 36, No. 15_suppl ( 2018-05-20), p. e24279-e24279
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2018
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Oncotarget, Impact Journals, LLC, Vol. 9, No. 33 ( 2018-05-01), p. 23289-23305
    Type of Medium: Online Resource
    ISSN: 1949-2553
    URL: Issue
    Language: English
    Publisher: Impact Journals, LLC
    Publication Date: 2018
    detail.hit.zdb_id: 2560162-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...