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
Filter
  • American Association for Cancer Research (AACR)  (10)
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 8_Supplement ( 2023-04-14), p. CT116-CT116
    Abstract: Background: The cytokine GDF15 is overexpressed in solid malignant tumors such as colorectal, lung and urothelial cancer, where it modulates T cells, dendritic cells (DCs) and myeloid-derived cells, driving the tumor microenvironment toward an immunosuppressive, tumor-promoting state. AZD8853 is a humanized immunoglobulin G1 monoclonal antibody that binds to, and neutralizes, GDF15. Anti-GDF15 treatment increased T cell proliferation and DC activation, leading to an antitumor immune response in preclinical studies of anti-PD-L1 resistant models. In vitro and in vivo preclinical data support the potential antitumor activity of AZD8853 in pts with selected advanced/metastatic cancers. Methods: This Phase I/IIa, first-in-human, open-label study (NCT05397171) assesses the safety, pharmacokinetics (PK), pharmacodynamics (PD) and preliminary efficacy of AZD8853 in pts with histologically or cytologically confirmed locally advanced, unresectable or metastatic mismatch repair-proficient colorectal cancer (pMMR-CRC), non-small-cell lung cancer (NSCLC) and urothelial carcinoma (UC). Up to 165 pts will be enrolled in 3 parts: Part A, dose escalation; Part B, pharmacodynamics expansion; and Part C, efficacy expansion. All pts receive AZD8853 IV. Eligible pts are ≥18 years old with ≥1 measurable target lesion per RECIST v1.1, ECOG PS of 0/1, life expectancy ≥12 weeks and adequate organ and bone marrow function. Pts with NSCLC must have had ≥1 prior line of systemic treatment in the advanced/metastatic setting, and no sensitizing EGFR or ALK aberrations. Pts with pMMR-CRC must have had ≥2 prior treatments in the advanced/metastatic setting. Pts with UC must have had ≥1 prior treatment in the advanced/metastatic setting including platinum-containing therapy and/or a PD-(L)1-inhibitor. Pts with Grade ≥2 unresolved toxicities from prior therapy, symptomatic CNS metastases or leptomeningeal disease, or prespecified active/ongoing infections are excluded. The primary objective is safety, including dose-limiting toxicities, adverse events (AEs), serious AEs and AEs leading to AZD8853 discontinuation. The secondary objectives include assessment of efficacy (objective response rate, disease control rate, duration of response, percentage change from baseline in target lesion size, change from baseline in circulating tumor DNA, and progression-free and overall survival), PK and immunogenicity. Changes in GDF15 serum levels are measured in Parts A and B. Tumoral CD8+ T cell infiltration is measured in a subset of pts from Part B using PET/CT imaging and IHC of paired biopsies. The study is currently recruiting at centers in the USA and Canada with additional sites planned in the UK, France and Spain. Citation Format: Benedito A. Carneiro, Maria Diab, Brian A. Van Tine, Anthony F. Shields, Albiruni Abdul Razak, John F. Hilton, Rafael Santana-Davila, Elhan Sanai, Jorge Zeron-Medina, Veronique Bragulat, Kath Lowery, Arthur Lambert, John Hood, Rakesh Kumar, Duncan Jodrell, Patricia LoRusso. First-in-human study of AZD8853, an anti-growth and differentiation factor 15 (GDF15) antibody, in patients (pts) with advanced/metastatic solid tumors [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 2 (Clinical Trials and Late-Breaking Research); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(8_Suppl):Abstract nr CT116.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 5467-5467
    Abstract: NSCLC accounts for approximately 85% of all lung cancers. Based on tumor histology, NSCLC can be subdivided into adenocarcinoma (AC), squamous cell carcinoma (SCC), and large cell carcinoma (LCC), which account for 40%, 25-30% and 10-15% of NSCLC cases, respectively. In a fraction of AC cases, driver oncogenes have been identified that enable effective treatment with targeted therapies. For most NSCLC cases, however, etiology is still unknown and effective targeted therapies have yet to be achieved. Additionally, even cancers that initially respond to targeted therapies eventually develop resistance, creating the need for other therapeutic approaches. EGFR is one of the best characterized driver oncogenes in NSCLC and also is frequently expressed at high levels in this indication. In house immunohistochemical analysis found EGFR to be highly expressed in 23.8% of AC (n = 21), 60% of SCC (n = 10) and 50% of LCC (n = 8), confirming published data that EGFR represents an attractive therapeutic target for NSCLC. As a novel approach to targeting EGFR-expressing tumors, we have developed IMGN289, an antibody-“drug” conjugate (ADC) consisting of the J2898A antibody (Ab), which selectively binds to EGFR and inhibits EGFR-driven tumor cell growth, conjugated to the maytansinoid DM1, a potent anti-microtubule agent, via the SMCC thioether linker. IMGN289 showed significantly enhanced cytotoxic activity, relative to cetuximab or unconjugated J2898A, against a panel of NSCLC cell lines dependent on EGFR signaling. In vitro, anti-EGFR Abs typically inhibit less than 70% of tumor cell growth at 30 nM concentration, whereas IMGN289 exposure approached 100% inhibition at a significantly lower concentration. Enhanced potency of IMGN289 against EGFR-dependent tumors was demonstrated in vivo in the H292 xenograft tumor model, where the minimally effective doses of IMGN289 and its J2898A Ab were 1 and 3 mg/kg, respectively. In addition, IMGN289 was effective against EGFR-expressing NSCLC cells that are not dependent on EGFR signaling and therefore resistant to anti-EGFR Abs. For example, IMGN289 was active against H226 and H1703 mesenchymal cell lines in vitro and H1703 xenograft tumor in vivo, while neither cetuximab nor J2898A alone was active. IMGN289 was also active against EGFR mutant HCC827 cell lines that have acquired resistance to EGFR tyrosine kinase inhibitors through T790M EGFR mutation or MET gene amplification. Thus, EGFR-targeting IMGN289 achieves a distinct anti-tumor mechanism that is independent of EGFR pathway inhibition. In summary, IMGN289 is highly active against EGFR-positive NSCLC cells regardless of dependency on the EGFR pathway and represents a promising therapeutic candidate for NSCLC. Citation Format: Thomas D. Chittenden, Yulius Y. Setiady, Peter U. Park, Jose F. Ponte, Ling Dong, Anna Skaletskaya, Christina N. Carrigan, Alfred A. Villaluz, Jan Pinkas, Robert J. Lutz, John M. Lambert. IMGN289, an EGFR-targeting antibody-maytansinoid conjugate with potent activity against non-small cell lung cancer (NSCLC) regardless of dependency on EGFR pathway. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 5467. doi:10.1158/1538-7445.AM2013-5467
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    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 ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2010
    In:  Cancer Research Vol. 70, No. 7 ( 2010-04-01), p. 2924-2931
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 7 ( 2010-04-01), p. 2924-2931
    Abstract: A subset of the mucosotropic human papillomaviruses (HPV), including HPV16, are etiologic agents for the vast majority of cervical cancers, other anogenital cancers, and a subset of head and neck squamous cell carcinomas. HPV16 encodes three oncogenes: E5, E6, and E7. Although E6 and E7 have been well-studied and clearly shown to be important contributors to these cancers, less is known about E5. In this study, we used E5 transgenic mice to investigate the role of E5 in cervical cancer. When treated for 6 months with estrogen, a cofactor for cervical carcinogenesis, E5 transgenic mice developed more severe neoplastic cervical disease than similarly treated nontransgenic mice, although no frank cancers were detected. In addition, E5 when combined with either E6 or E7 induced more severe neoplastic disease than seen in mice expressing only one viral oncogene. Prolonged treatment of E5 transgenic mice with exogenous estrogen uncovered an ability of E5 to cause frank cancer. These data indicate that E5 acts as an oncogene in the reproductive tracts of female mice. Cancer Res; 70(7); 2924–31
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2010
    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. 71, No. 8_Supplement ( 2011-04-15), p. 4565-4565
    Abstract: CD37 is a B-cell surface antigen that is an attractive target for antibody and antibody-drug conjugate mediated therapies due to its restricted expression profile. It is expressed on malignant B-cells in NHL and CLL, but on normal tissue its expression is highly restricted to B-cells present in blood and lymphoid tissues. A large panel of anti-CD37 murine monoclonal antibodies were generated and screened for their specific CD37 binding affinity, direct anti-proliferative activity and pro-apoptotic activity against lymphoma cell lines. Selected antibodies were humanized by variable domain resurfacing and one antibody, designated K7153A, demonstrated the best overall activity in terms of direct antibody activity as well as effector function. K7153A demonstrated much stronger pro-apoptotic activity against Ramos and Raji cells than either of two reference compounds, the anti-CD37 SMIP TRU-016 or the anti-CD20 antibody rituximab, and did not require cross-linking to achieve this effect. The antibody-maytansinoid conjugate, IMGN529, was produced by conjugation of K7153A with the potent maytansinoid, DM1, via the non-cleavable linker, SMCC. IMGN529 retains the high specific binding affinity of the K7153A antibody, with an EC50 of 0.5 nM. IMGN529 also demonstrated the same strong pro-apoptotic activity as the K7153A antibody against Ramos cells, with an EC50 of 0.1 nM. Antibody-dependent cell-mediated cytotoxicity (ADCC) assays, using purified human NK cells as effector cells, showed that K7153A and IMGN529 have similar potent ADCC activity against Ramos and Daudi cells with an EC50 of less than 10 pM. In addition, both K7153A and IMGN529 demonstrated comparable complement-dependent cytotoxicity (CDC) in the presence of human complement against Ramos cells. These results indicate that IMGN529 retains the intrinsic functions of the K7153A antibody. IMGN529 was highly cytotoxic in vitro against NHL cell lines such as Daudi, BJAB, Namalwa and SU-DHL-4 with a greater degree of cell killing and lower EC50 value (19 – 36 pM) than the K7153 antibody alone. In contrast, TRU-016 showed no effect on any of these cell lines and rituximab was only active against SU-DHL-4 cells. In vivo, IMGN529 showed markedly higher efficacy against established SU-DHL-4 and BJAB xenograft tumors than the K7153A antibody alone, with significant anti-tumor activity at single doses of 5 mg/kg or lower. Together, these results demonstrate that IMGN529 combines the strong pro-apoptotic activity, CDC and ADCC activity of its anti-CD37 antibody component with the potent cytotoxic activity provided by the targeted delivery of its maytansinoid payload. IMGN529 is a highly active antibody-drug conjugate with a unique combination of anti-tumor activities and is therefore a promising therapeutic candidate for the treatment of CD37-positive lymphomas and leukemias. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 4565. doi:10.1158/1538-7445.AM2011-4565
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    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: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 4531-4531
    Abstract: Antibody-drug conjugates (ADCs) are being actively pursued as a new modality to treat cancer following the regulatory approval of Adcetris@ and Kadcyla@. ADCs consist of a cytotoxic agent, or drug, conjugated to a targeting antibody (Ab) through a linker. The two approved ADCs (and most ADCs now in the clinic) are heterogeneous conjugates with an average molar drug to Ab ratio (DAR) of 3-4 (potentially ranging from 0-8 for individual molecules). To understand the effects of different DAR ranges on the preclinical properties of ADCs using a maytansinoid cytotoxic agent, we prepared a series of conjugates with a cleavable linker (M9346A-sulfo-SPDB-DM4 targeting folate receptor alpha) or an uncleavable linker (J2898A-SMCC-DM1 targeting epidermal growth factor receptor) with varying DAR, and evaluated their biochemical characteristics, in vivo stability, efficacy, and tolerability. Both M9346A-sulfo-SPDB-DM4 and J2898A-SMCC-DM1 conjugates with low (average ∼2, range 0-4) to very high DAR (average 10, range 7-14) were prepared in good yield, high monomer content and low free drug levels. At constant Ab concentration, the in vitro potency consistently increased with increasing DAR. We then characterized the in vivo disposition of these ADCs. First, pharmacokinetic analysis showed that conjugates with an average DAR below ∼6 had comparable clearance rates, but at an average DAR around 9-10 rapid clearance was observed. Biodistribution studies in mice showed that these 9-10 DAR conjugates rapidly distribute to the liver, with the maximum%ID/g for this organ at 24-28% compared with 7-10% for lower DAR conjugates (all at 2-6 h post-injection). We further studied the efficacy of these ADCs in mouse xenograft models at both constant antibody and constant maytansinoid doses. At constant Ab dose, conjugates with average DAR ∼6 are more efficacious than conjugates with DAR ∼3. However, DAR 9-10 conjugates are either less or similarly active compared to DAR 6 conjugates, consistent with the observed rapid clearance. At constant maytansinoid dose, conjugates with 2 - 6 DAR show similar efficacy, while those with 9-10 DAR were again less active. Tolerability studies monitoring body weight loss show that conjugates with different DAR, even as high as 9-10, have comparable tolerability based on total administered maytansinoid dose. In summary, our preclinical findings on tolerability and efficacy suggest that maytansinoid conjugates with DAR in the range between 2 - 6 have better therapeutic index than conjugates with very high DAR (∼9-10). These very high DAR ADCs suffer from decreased efficacy likely due to faster clearance. These results support the use of DAR 3-4 for maytansinoid ADCs, but suggest the exploration of lower or higher DAR depending on the biology of the target antigen. Citation Format: Xiuxia Sun, Jose F. Ponte, Nicholas C. Yoder, Jennifer Coccia, Leanne Lanieri, Rassol Laleau, Qifeng Qiu, Rui Wu, Erica Hong, Megan Bogalhas, Lintao Wang, Erin K. Maloney, Olga Ab, Hans K. Erickson, Thomas A. Keating, Ravi Chari, John M. Lambert. Effects of drug load on therapeutic index for antibody-maytansinoid conjugates. [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 4531. doi:10.1158/1538-7445.AM2015-4531
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 17, No. 1_Supplement ( 2018-01-01), p. LB-A01-LB-A01
    Abstract: Introduction: Although human Papillomavirus (HPV) has been identified as a causative reagent for cervical cancer (CVC), accumulating evidence indicates that HPV alone is insufficient to induce CVC. The molecular mechanisms underlying CVC development are largely unknown. Our previous in vitro studies indicated that YAP1, the major effector of the Hippo pathway, interacts with HPV to drive the progression of CVC. However, whether the Hippo/YAP1 pathway also regulates cervical carcinogenesis in vivo is unknown. Aims: To examine the molecular mechanism(s) by which the Hippo/YAP pathway interacts with HPV to regulate the cervical carcinogenesis. Methods: The cBioportal for Cancer Genomics, an online tool developed to visualize The Cancer Genomic Atlas (TCGA) studies, was used to analyze alterations of the Hippo pathway in human CVC. Primary cervical epithelial cells and cervical cancer cell lines were used to determine effects of YAP1 on cell susceptibility to HPV infection. KRT14-YAPS127A mice, which express constitutively active YAP1 (YAPS127A) in cervical epithelial cells under doxycycline (Dox) induction, and KRT14-E6/E7-YAPS127A mice, which express both HPV E6/E7 and YAPS127A in cervical epithelial cells under Dox induction, were used to examine the role of YAP1 gene in cervical carcinogenesis. Results: Multidimensional cancer genomic data showed that disruption of the Hippo signaling pathway is a common event in human CVC, which is featured by frequent amplification and up-regulation of oncogenes and deletion/mutation of tumor suppressors. Moreover, the disrupted Hippo pathway is associated with poor prognosis of CVC patients. Dox-induced expression of YAPS127A in KRT14-YAPS127A mouse cervical epithelial cells resulted in neoplasia of cervical epithelium within 2 months. Most KRT14-YAPS127A mice developed invasive CVC after induction with dox for 6 months. Metastatic spread of CVC induced urethral and outlet obstruction in some KRT14-YAPS127A mice. Around 50% of KRT14-YAPS127A mice died of CVC within 16 months of Dox induction. Histological analysis revealed that KRT14-YAPS127A mice developed squamous cell carcinomas (SCC). Although KRT14-E6/E7 mice (only expressing E6/E7) rarely developed CVC after induction with Dox for 12 months, KRT14-E6/E7-YAPS127A mice developed cervical cancer within 4 months of Dox induction. Compared to KRT14-YAPS127A mice, cancer cells in the cervical tissue of KRT14-E6/E7-YAPS127A mice were more proliferative and invasive. Mechanistic studies showed that ectopic expression of YAPS127A in human cervical epithelial cells resulted in a significant increase in cell susceptibility to HPV infection, while inhibition of YAP1 activity reduced the acceptability of cervical epithelial cells to HPV infection. Intriguingly, we found that activation of YAP induced up-regulation of the putative HPV cellular receptor and disrupted innate immunity in cervical epithelial cells. Conclusion: Our research demonstrates that constitutive activation of YAP1 is sufficient to induce cervical SCC. Hyper-activation of YAP increases the susceptibility of cervical epithelial cells to HPV infection. HPV, in turn, synergistically promotes YAP1-induced initiation and progression of cervical cancer. The Hippo/YAP signaling pathway may represent a promising target for developing novel strategies to prevent and treat cervical cancer. [C.H., X.L., and C.H. contributed equally to this work.] Citation Format: Chunbo He, Xiangmin Lv, Cong Huang, Guohua Hua, Jin Zhou, Zhengfeng Wang, Jixin Dong, Peter C Angeletti, Paul F. Lambert, Bo R Rueda, John S Davis, Cheng Wang. Yes-associated protein 1 regulates cervical cancer initiation and progression via suppressing innate immunity and increasing cell susceptibility to human papillomavirus infection [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 LB-A01.
    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 ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2010
    In:  Cancer Research Vol. 70, No. 8_Supplement ( 2010-04-15), p. 3668-3668
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 8_Supplement ( 2010-04-15), p. 3668-3668
    Abstract: Background: Tamoxifen is effective in the treatment of estrogen receptor alpha (ER)-positive breast cancer. However, almost all responding patients develop resistance. Understanding the mechanisms leading to resistance is crucial for the development of more efficient therapies. In a functional screen for genes causing tamoxifen resistance, we have identified the breast cancer antiestrogen resistance 4 (BCAR4) gene. Forced expression of BCAR4 in estrogen-dependent breast cancer cell lines induced antiestrogen resistance. Objective: We explored whether BCAR4 mRNA levels in primary tumors predict tamoxifen resistance and/or tumor aggressiveness, and searched for signaling pathways involved in BCAR4-induced resistance to tamoxifen. Experimental design: BCAR4 mRNA levels were measured by quantitative RT-PCR. In 280 ER-positive primary tumors from patients receiving tamoxifen as first-line monotherapy for advanced disease, the associations between BCAR4 mRNA levels and progression-free survival (PFS) and clinical benefit were evaluated. BCAR4 mRNA levels in primary tumors from 506 patients with lymph node-negative, ER-positive cancer were evaluated for their association with distant metastasis-free survival (MFS). None of them received systemic adjuvant therapy, thereby preventing confounding effects of systemic treatment. In order to study BCAR4 function, we forced the expression of BCAR4 in estrogen-dependent ZR-75-1 breast cancer cells, and analyzed changes in phospho-protein expression. Results: In the study for the association of BCAR4 with tamoxifen resistance, high BCAR4 mRNA levels were significantly associated with a shorter PFS and poor clinical benefit (measurable tumor response or no change & gt; 6 months). Multivariate analysis including the traditional predictive factors indicated that a high BCAR4 mRNA level is an independent predictive factor for PFS. In the assessment of tumor aggressiveness, high BCAR4 mRNA levels were associated with a shorter MFS and overall survival (OS). Multivariate analyses showed that a high BCAR4 mRNA level was an independent prognostic factor for MFS and OS. Forced expression of BCAR4 induced phosphorylation of ERBB2, ERBB3, and their down-stream mediators ERK1/2 and AKT in ZR-75-1 cells. Proliferation of these cells was inhibited by the knockdown of ERBB2 or ERBB3 with specific siRNAs, confirming the role of these receptors in BCAR4-induced tamoxifen resistance. The knockdown of ER had no effect on the proliferation of BCAR4-expressing cells, indicating that this mechanism of resistance is independent of ER signaling. Conclusions: Our data indicate that high BCAR4 mRNA levels predict resistance to tamoxifen treatment and poor outcome. The results of our experimental cell model also suggest that tumors expressing BCAR4 may rely on ERBB2/ERBB3 signaling. Therefore, patients with tumors expressing BCAR4 may be eligible to receive ERBB-targeted therapy. Note: This abstract was not presented at the AACR 101st Annual Meeting 2010 because the presenter was unable to attend. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 3668.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2010
    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 ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 5463-5463
    Abstract: EGFR is an attractive target for the treatment of a variety of solid tumors because of its role as a driver oncogene and high level of expression. Four EGFR-targeting agents, including two antibodies (Abs), have been approved for clinical use. Despite anti-tumor benefits, inhibition of EGFR pathway is associated with significant dermatologic toxicities; resistance to EGFR antagonists also develops. To enhance potency with comparable or better tolerability, we developed IMGN289, an EGFR-targeting antibody-“drug” conjugate (ADC) that disrupts tumor growth both by inhibiting EGFR signaling and through direct anti-mitotic activity. To reduce potential dermatologic toxicities associated with EGFR pathway inhibition, a unique Ab discovery approach was employed. Hybridomas from mice immunized with EGFR-expressing tumor cells were screened for EGFR binding and selective inhibitory activity against EGFR-dependent tumor cells. This approach revealed a novel class of Ab with selective EGFR antagonistic activity. A humanized lead Ab was identified, J2898A, which was comparable in potency to cetuximab in vitro against a panel of EGFR-dependent tumor cell lines and in vivo against two head and neck tumor xenograft models. Notably, in cultures of human primary keratinocytes, this Ab was markedly less cytotoxic than cetuximab and did not affect TNFα-induced cytokine production, which has been implicated in chronic dermatologic toxicities induced by other anti-EGFR agents. To further enhance cytotoxic activity and to potentially overcome resistance to EGFR-targeting therapies, J2898A was conjugated to the maytansinoid DM1, a potent anti-tubulin agent, via a non-cleavable linker, SMCC. IMGN289 was not only more potent than J2898A against EGFR-dependent tumors, but also was effective against EGFR-positive tumor cells that grow independently of signaling via the EGFR pathway or have acquired resistance to EGFR inhibitors, including lung adenocarcinoma cell lines harboring the T790M EGFR mutation or MET gene amplification. Despite having potent activity against EGFR-expressing tumor cells, IMGN289 was less toxic to cultured keratinocytes than cetuximab. Moreover, a toxicology study in cynomolgus monkeys demonstrated that IMGN289 was well tolerated and exhibited a similar toxicity profile to that published for trastuzumab emtansine (T-DM1), another ADC which utilizes SMCC-DM1 as the selected linker-payload format. In summary, IMGN289 combines EGFR inhibition mediated by its J2898A Ab component with the potent cytotoxicity provided by its DM1 payload, and is highly active against EGFR-positive tumors regardless of their dependency on the EGFR pathway. IMGN289 thus represents a promising novel candidate for treatment of EGFR-expressing solid tumors. Citation Format: Yulius Y. Setiady, Peter U. Park, Jose F. Ponte, Ling Dong, Anna Skaletskaya, Jennifer A. Coccia, Erica Hong, Lauren Clancy, Lingyun Rui, Jan Pinkas, Robert J. Lutz, John M. Lambert, Thomas D. Chittenden. Development of a novel antibody-maytansinoid conjugate, IMGN289, for the treatment of EGFR-expressing solid tumors. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 5463. doi:10.1158/1538-7445.AM2013-5463
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    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 ...
  • 9
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 15, No. 8 ( 2016-08-01), p. 1870-1878
    Abstract: The promise of tumor-selective delivery of cytotoxic agents in the form of antibody–drug conjugates (ADC) has now been realized, evidenced by the approval of two ADCs, both of which incorporate highly cytotoxic tubulin-interacting agents, for cancer therapy. An ongoing challenge remains in identifying potent agents with alternative mechanisms of cell killing that can provide ADCs with high therapeutic indices and favorable tolerability. Here, we describe the development of a new class of potent DNA alkylating agents that meets these objectives. Through chemical design, we changed the mechanism of action of our novel DNA cross-linking agent to a monofunctional DNA alkylator. This modification, coupled with linker optimization, generated ADCs that were well tolerated in mice and demonstrated robust antitumor activity in multiple tumor models at doses 1.5% to 3.5% of maximally tolerated levels. These properties underscore the considerable potential of these purpose-created, unique DNA-interacting conjugates for broadening the clinical application of ADC technology. Mol Cancer Ther; 15(8); 1870–8. ©2016 AACR.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2007
    In:  Cancer Research Vol. 67, No. 13 ( 2007-07-01), p. 6106-6112
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 67, No. 13 ( 2007-07-01), p. 6106-6112
    Abstract: High-risk human papillomaviruses (HPVs), which cause the vast majority of cervical cancer, other anogenital cancers, and a subset of head and neck squamous cell carcinomas, encode three oncogenes: E5, E6, and E7. To determine the oncogenic properties of HPV16 E5 in vivo, we previously generated K14E5 transgenic mice, in which expression of E5 was directed to the basal compartment of stratified squamous epithelia. In these mice, E5 induced epidermal hyperplasia and spontaneous skin tumors. In the current study, we determined how E5 contributes to tumor formation in the skin using a multistage model for skin carcinogenesis that specifies the role of genes in three stages: initiation, promotion, and malignant progression. Both initiation and promotion are required steps for papilloma formation. K14E5 mice treated with the initiating agent 7,12-dimethylbenz(a)anthracene (DMBA) developed more papillomas than like-treated nontransgenic mice, whereas neither K14E5 nor nontransgenic mice treated with the promoting agent 12-O-tetradecanoylphorbol-13-acetate (TPA) developed papillomas. K14E5 mice treated with both DMBA and TPA to induce large numbers of papillomas had a higher incidence and earlier onset of carcinoma progression compared with like-treated nontransgenic mice. Thus, HPV16 E5 contributes to two stages of skin carcinogenesis: promotion and progression. The progressive neoplastic disease in K14E5 mice differed from that in nontransgenic mice in that benign tumors converted from exophytic to endophytic papillomas before progressing to carcinomas. Initial genetic and immunohistopathologic analyses did not determine the underlying basis for this distinct morphology, which correlates with a highly penetrant neoplastic phenotype. [Cancer Res 2007;67(13):6106–7]
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2007
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...