feed icon rss

Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Modern Pathology, Elsevier BV, Vol. 36, No. 8 ( 2023-08), p. 100192-
    Type of Medium: Online Resource
    ISSN: 0893-3952
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2023
    detail.hit.zdb_id: 2041318-X
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    AME Publishing Company ; 2018
    In:  Translational Lung Cancer Research Vol. 7, No. 5 ( 2018-12), p. 716-721
    In: Translational Lung Cancer Research, AME Publishing Company, Vol. 7, No. 5 ( 2018-12), p. 716-721
    Type of Medium: Online Resource
    ISSN: 2218-6751 , 2226-4477
    Language: Unknown
    Publisher: AME Publishing Company
    Publication Date: 2018
    detail.hit.zdb_id: 2754335-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    Springer Science and Business Media LLC ; 2022
    In:  Clinical Proteomics Vol. 19, No. 1 ( 2022-12)
    In: Clinical Proteomics, Springer Science and Business Media LLC, Vol. 19, No. 1 ( 2022-12)
    Abstract: Breast cancer (BC) is the most common cancer and among the leading causes of cancer death in women. It is a heterogeneous group of tumours with numerous morphological and molecular subtypes, making predictions of disease evolution and patient outcomes difficult. Therefore, biomarkers are needed to help clinicians choose the best treatment for each patient. For the last years, studies have increasingly focused on biomarkers obtainable by liquid biopsy. Circulating proteins (from serum or plasma) can be used for inexpensive and minimally invasive determination of disease risk, early diagnosis, treatment adjusting, prognostication and disease progression monitoring. We provide here a review of the main published studies on serum proteins in breast cancer and elaborate on the potential of circulating proteins to be predictive and/or prognostic biomarkers in breast cancer.
    Type of Medium: Online Resource
    ISSN: 1542-6416 , 1559-0275
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2022
    detail.hit.zdb_id: 2163624-2
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2014
    In:  Cancer Research Vol. 74, No. 19_Supplement ( 2014-10-01), p. NG01-NG01
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. NG01-NG01
    Abstract: Triple negative breast cancer (TNBC) is clinically defined by lack of expression of estrogen and progesterone receptors and no overexpression/amplification of human epidermal growth factor receptor 2 (HER2). It affects approximately one-fourth of breast cancer patients and is usually characterized by an aggressive phenotype with high mitotic index and poor prognosis. No TNBC-targeted therapeutic exists and the standard of care is currently limited to cytotoxic chemotherapy. Overexpression of human epidermal growth factor receptor (EGFR) as well as inactivation or decreased expression of phosphatase and tensin homolog deleted on chromosome 10 (PTEN) are frequent in TNBC. Thus, inhibition of EGFR and/or the PI3K/Akt pathway are attractive therapeutic strategies for this malignancy. Preclinical evidence, however, suggests that inhibition of the PI3K/Akt axis induces compensatory expression and activation of upstream receptor tyrosine kinases, including EGFR and, most prominently, human epidermal growth factor receptor 3 (HER3). This may reduce the antitumor effects of single-agent PI3K pathway blockade. Furthermore, studies using in vitro models of resistance to the anti-EGFR antibody cetuximab suggest that HER3 expression itself can limit the sensitivity to this agent by increasing EGFR/HER3 heterodimerization and activation of downstream pathways. In this work we hypothesized that targeting both EGFR and HER3, in combination with PI3K/Akt inhibition, would result in enhanced therapeutic activity in EGFR-positive TNBC. We treated PTEN-negative EGFR-positive HCC70 and MDA-MB-468 TNBC cell lines with GDC-0068, a selective inhibitor of the Akt 1/2/3 isoforms, GDC-0941, a class I selective pan-PI3K inhibitor, MEHD7945A, an antibody targeting both EGFR and HER3 and the combinations of these inhibitors. Compared to single agents, the combination of GDC-0068 or GDC-0941 and MEHD7945A prevented the induction of EGFR and HER3 phosphorylation, enhanced the inhibition of the PI3K and ERK downstream pathways and resulted in superior inhibition of cell proliferation/viability compared to single agents. To expand our findings in vivo, we first tested the efficacy of MEHD7945A in combination with either the Akt or PI3K inhibitor in both MDA-MB-468- and HCC70-derived xenografts. While the tumors responded only modestly to single agent GDC-0068, GDC-0941 and MEHD7945A, the combination of GDC-0068 or GDC-0941 with MEHD7945A yielded superior tumor growth inhibition compared to monotherapies. We next investigated the levels of EGFR and HER3 expression/activation in HCC70 tumors collected at the end of the experiments by reverse-phase protein detection. As expected, increase of both EGFR and HER3 expression and phosphorylation induced by Akt or PI3K was prevented by the addition of MEHD7945A. We then tested the activity of the same treatments in a patient-derived xenograft (PDX) model of TNBC available in our laboratory. These tumors were characterized by undetectable levels of PTEN, high levels of EGFR and ∼70% staining for Ki67. When used as single agents, GDC-0068, GDC-0941 and MEHD7945A delayed tumor growth. However, their combinations elicited durable tumor stasis. Consistently with the cell-based xenografts, MEHD7945A counteracted EGFR and HER3 phosphorylation caused by PI3K/Akt inhibition. Collectively, these data show that targeting both EGFR and HER3 enhances the antitumor effects of PI3K/Akt inhibitors. In order to dissect the role of HER3 inhibition in these models, we compared the activity of cetuximab with MEHD7945A in combination with either GDC-0068 or GDC-0941 in HCC70 cells. Both antibodies enhanced the antiproliferative activity mediated by PI3K/Akt inhibition in cells stimulated with the EGFR ligand epidermal growth factor; however MEHD7945A was superior to cetuximab in cooperating with the antiproliferative activity of GDC-0068 and GDC-0941 in cells stimulated with the HER3 ligand heregulin. We next compared the antitumor activity of cetuximab and MEHD7945A in combination with GDC-0941 in HCC70-derived xenografts. While concomitant targeting of EGFR, HER3 and PI3K was superior to monotherapies and led to tumor shrinkage, the combination of cetuximab and GDC-0941 did not result to any further inhibition of tumor growth compared to single agent treatments. Biochemically, both MEHD7945A and cetuximab blocked EGFR phosphorylation in these xenografts; however, only MEHD7945A decreased HER3 activation. These results confirm that HER3 plays an important role in limiting the efficacy of PI3K inhibition in this setting. To investigate whether changes in EGFR and HER3 levels can affect the response to anti-EGFR therapy in TNBC patients, we measured the expression of these receptors in samples from patients enrolled in two pilot neoadjuvant clinical trials testing the antitumor activity of the anti-EGFR antibodies panitumumab (40 patients) and cetuximab (30 patients) in combination with standard chemotherapy. Of 40 patients treated with panitumumab-based therapy 19 (47.5%) achieved pathological complete response (pCR) after 24 weeks and 21 (52.5%) showed residual disease at the time of surgery. We measured EGFR and HER3 expression by immunohistochemistry in the 40 pre-treatment specimens and in the 21 residual invasive tumors excised at surgery. Patients with higher baseline EGFR expression were more likely to achieve pCR. Moreover, EGFR levels were decreased in the residual tumors of nine out of 21 non-pCR patients when compared to the paired baseline specimens (p=0.07). HER3 staining, available for 13 non-responder patients, showed higher HER3 expression in the residual lesions of 7 out of 13 non-pCR patients compared to the paired pre-treatment samples (p=0.01). Of 30 patients enrolled in the study testing the antitumor activity of cetuximab-based therapy, 9 experienced pCR. Consistently, HER3 expression was found upregulated in the residual tumors of 11 out of 19 non-pCR patients when compared to the baseline paired specimens (p=0.103). These results suggest that high EGFR expression may be required for optimal response to anti-EGFR therapeutic antibodies and that HER3 expression increases following anti-EGFR therapy in patients that do not experience complete tumor regression. In this work we provide evidence that HER3 plays a potential role in limiting the antitumor activity of both PI3K/Akt inhibitors and anti-EGFR agents. We demonstrate that simultaneous targeting of EGFR and HER3 is required to enhance the efficacy of PI3K/Akt inhibition in preclinical models of EGFR-positive TNBC. Further, our clinical findings suggest that HER3 expression is induced in TNBC patients with a lower probability of achieving tumor remission upon anti-EGFR therapy. Therefore, the rationale behind targeting simultaneously EGFR, HER3 and PI3K in TNBC is based on: 1) nearly half of TNBCs express high levels of EGFR, 2) TNBCs often express low levels of PTEN and 3) HER3 upregulation may limit their sensitivity to targeted therapy. In addition, tumor cells that express relatively high levels of these receptors can function as molecular “flags” for immune-mediated antibody dependent cytotoxicity, intrinsic characteristics of IgG type I antibodies such as MEHD7945A in vivo. Given that both pharmacokinetic and pharmacodynamic data for MEHD7945A, GDC-0068 and GDC-0941 are already available, the design of phase II clinical trials testing the activity of these possible combinations in TNBC would be straightforward. Patient enrollment may be based on high EGFR expression with the requirement of mandatory “on-treatment” biopsies to evaluate both pathway inhibition and possible HER3 upregulation upon therapy. In conclusion, we believe that simultaneous inhibition of EGFR, HER3 and the PI3K/Akt pathway has the potential to greatly expand the percentage of TNBC patients who can benefit from targeted therapy. Citation Format: Maurizio Scaltriti, Jessica Tao, Pau Castel, Nina Radosevic-Robin, Moshe Elkabets, Neil Auricchio, Dejan Juric, Frédérique Penault-Llorca, Jose Baselga. Blockade of EGFR and HER3 enhances PI3K/Akt antitumor activity in triple negative breast cancer. [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 NG01. doi:10.1158/1538-7445.AM2014-NG01
    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 ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 550-550
    Abstract: Background. Triple negative breast cancers (TNBC) represent about 15 % of invasive breast carcinomas. These tumors are identified in clinical practice according to their “triple negative” immunophenotype (estrogen receptor, progesterone receptor and HER2 negative). TNBC show aggressive behavior and an unusual sarcoma-like pattern of metastasis. Identification of predictive biomarkers is critical to optimal therapeutic management of these tumors. TNBC are characterized by increased genomic instability. These tumors present complex pattern of numerous low-amplitude genomic gains and losses as well as shorter telomeres. Aim. Identification of predictive biomarkers of response to neoadjuvant chemotherapy in TNBC by evaluating telomeric parameters and genomic instability. Methods. We studied 38 TNBC patients treated with neoadjuvant chemotherapy according to the TVA protocol (FEC 100, docetaxel, panitumumab). DNA and RNA were extracted from snap-frozen tumor samples obtained before neoadjuvant therapy. The pathological response to treatment was evaluated according to Chevallier's and Sataloff's classifications and correlated with aCGH profiles (8x60K, Agilent) and telomere characteristics, including telomere length (qPCR) and expression (qRT-PCR) of telomerase (hTERT) and shelterin complex genes (TRF1, TRF2, POT1, TPP1, RAP1 and TIN2). Results. Telomere shortening and high hTERT expression were found in more aggressive tumors according to TNM classification (P & lt; 0.04), and hTERT expression was also highly correlated with an increased tumor size (P= 0.0058). Short telomeres were strongly associated with incomplete histological response to neoadjuvant therapy according to Chevallier's (P= 0.0004) and Sataloff's classifications, (P= 0.0007). hTERT expression was significantly increased in cases with incomplete response (P & lt; 0.04). Among shelterin complex genes, TRF2 and TPP1 overexpression was found in more aggressive TNBC (according to TNM classification and SBR grading, P & lt; 0.05), but there was no correlation with pathological response. aCGH data indicated that genomic instability in non-responders was mainly represented by genomic gains. In particular, cases with incomplete therapeutic responses showed significantly larger size of gains than cases with complete responses. (P= 0.0089). Short telomeres were also associated with larger gains of genomic material (P= 0.0067). Of note, telomere shortening has been previously described as one of the mechanisms of genomic gains and amplifications. Conclusions. Telomere shortening and hTERT overexpression were observed in resistant TNBC. Genomic profile of non-responding tumors was characterized by larger genomic gains. Telomere length, hTERT expression and genomic instability profiles may represent predictive biomarkers of response to neoadjuvant chemotherapy in TNBC. Citation Format: Mathilde Gay-bellile, Nina Radosevic-Robin, Eleonore Eymard-Pierre, Fabrice Kwiatkowski, Marie-Mélanie Dauplat, Maud Privat, Catherine Abrial, Patricia Combes, Gwendoline Soler, Yves-Jean Bignon, Jean-Marc Nabholtz, Philippe Vago, Frédérique Penault-Llorca, Andreï Tchirkov. Genomic instability and telomere characteristics as predictive biomarkers of therapeutic response in triple-negative breast cancer. [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 550. doi:10.1158/1538-7445.AM2014-550
    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 ...
  • 6
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 23, No. 11 ( 2017-06-01), p. 2806-2816
    Abstract: Purpose: AXL receptor tyrosine kinase has been described as a relevant molecular marker and a key player in invasiveness, especially in triple-negative breast cancer (TNBC). Experimental Design: We evaluate the antitumor efficacy of the anti-AXL monoclonal antibody 20G7-D9 in several TNBC cell xenografts or patient-derived xenograft (PDX) models and decipher the underlying mechanisms. In a dataset of 254 basal-like breast cancer samples, genes correlated with AXL expression are enriched in EMT, migration, and invasion signaling pathways. Results: Treatment with 20G7-D9 inhibited tumor growth and bone metastasis formation in AXL-positive TNBC cell xenografts or PDX, but not in AXL-negative PDX, highlighting AXL role in cancer growth and invasion. In vitro stimulation of AXL-positive cancer cells by its ligand GAS6 induced the expression of several EMT-associated genes (SNAIL, SLUG, and VIM) through an intracellular signaling implicating the transcription factor FRA-1, important in cell invasion and plasticity, and increased their migration/invasion capacity. 20G7-D9 induced AXL degradation and inhibited all AXL/GAS6–dependent cell signaling implicated in EMT and in cell migration/invasion. Conclusions: The anti-AXL antibody 20G7-D9 represents a promising therapeutic strategy in TNBC with mesenchymal features by inhibiting AXL-dependent EMT, tumor growth, and metastasis formation. Clin Cancer Res; 23(11); 2806–16. ©2016 AACR.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    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. 1819-1819
    Abstract: Background: We have recently demonstrated (AACR 2013, abstract 4669) that antibody (Ab)-based EGFR targeting, sequentially combined with cytotoxic agents, can achieve pathologic complete response in almost 50% of triple-negative breast cancer (TNBC) patients (pts). However, the pts left with a residual tumor (RT), need a modification of treatment approach. We investigated whether expression of EGFR and two of its major cooperators, HER3 and MET, can offer a simple and practical base for neoadjuvant therapy (NAT) improvement in TNBC. Methods: The study included pts with a post-NAT RT in 2 pilot clinical trials of anti-EGFR Abs in TNBC, conducted from 2009 to 2011 by our team. One trial combined panitumumab with standard fluorouracil-epirubicin-cyclophosphamide and docetaxel (P-FEC-T) while another applied cetuximab with docetaxel (C-T). EGFR, HER3 and MET were assessed by immunohistochemistry (IHC) on pre- and post-NAT BC tissue. Expressions were evaluated by Quick score (% of positive cells x stain intensity as 0, 0.5, 1, 1.5, 2, 2.5 or 3; score range 0-300). At least 30 points of difference qualified the post-NAT score as changed, compared to the pre-NAT level. Results: 44 pts were eligible (P-FEC-T: 26 pts, C-T: 18 pts). Using 150 (the middle of the score range) as cut-off, we separated the tumors in each study on pre-NAT EGFR-high (score ≥ 150) and EGFR-low (score & lt; 150). The P-FEC-T trial had 13 EGFR-high tumors (median score 220) and 13 EGFR-low ones (median score 0). After NAT, the pre-NAT EGFR-high BC most frequently showed EGFR decrease (9/13), HER3 increase (6/10) and MET decrease (7/12). However, the pre-NAT EGFR-low tumors showed no change in EGFR (7/13) and HER3 (6/11) post-NAT, but most of them increased MET (7/11). The C-T trial had 7 EGFR-high (median score 210) and 11 EGFR-low cases (median score 70). The EGFR-high BC from that trial showed a similar pattern of score changes as the same group from the P-FEC-T trial: frequent EGFR decrease (4/7), HER3 increase (5/7) and MET decrease (4/7). The EGFR-low BC were more heterogeneous than in P-FEC-T: most frequently EGFR, HER3 and MET were increased (4/11, 5/11, 5/11 respectively) or unchanged (4/11, 4/11, 3/11 resp.). Conclusion: Our results indicate that: a) there is a TNBC subgroup with relatively high EGFR expression (score ≥ 150, median around 200) which resist to anti-EGFR treatment mainly by downregulating EGFR and upregulating HER3; b) TNBC with low EGFR expression (score & lt; 150) are a heterogeneous subgroup, which will frequently upregulate HER3 and/or MET after exposure to the anti-EGFR agents. The IHC assessment of EGFR, HER3 and MET before and on-NAT can help improving TNBC treatment by introduction of currently available anti-EGFR/HER3, anti-HER3 and anti-MET agents. Citation Format: Nina Radosevic-Robin, Catherine Abrial, Marie-Melanie Dauplat, Matthieu Roche, Anne Cayre, Maud Privat, Fabrice Kwiatkowski, Nassera Chalabi, Marie-Ange Mouret-Reynier, Yves-Jean Bignon, Philippe Chollet, Jean-Marc Nabholtz, Frederique Penault-Llorca. Heterogeneity of triple-negative breast cancer response to neoadjuvant treatment: tumor EGFR, HER3 and MET expressions can provide clues for therapy tailoring. [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 1819. doi:10.1158/1538-7445.AM2014-1819
    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: The Lancet Oncology, Elsevier BV, Vol. 22, No. 1 ( 2021-01), p. e18-e28
    Type of Medium: Online Resource
    ISSN: 1470-2045
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2021
    detail.hit.zdb_id: 2049730-1
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: npj Breast Cancer, Springer Science and Business Media LLC, Vol. 7, No. 1 ( 2021-09-17)
    Abstract: To find metastatic recurrence biomarkers of triple-negative breast cancer (TNBC) treated by neoadjuvant chemotherapy and anti-EGFR antibodies (NAT), we evaluated tumor genomic, transcriptomic, and immune features, using MSK-IMPACT assay, gene arrays, Nanostring technology, and TIL assessment on H & E. Six patients experienced a rapid fatal recurrence (RR) and other 6 had later non-fatal recurrences (LR). Before NAT, RR had low expression of 6 MHC class I and 13 MHC class II genes but were enriched in upregulated genes involved in the cell cycle-related pathways. Their TIL number before NAT in RR was very low ( 〈 5%) and did not increase after treatment. In post-NAT residual tumors, RR cases showed high expression of SOX2 and CXCR4 . Our results indicate that high expression of cell cycle genes, combined with cold immunological phenotype, may predict strong TNBC resistance to NAT and rapid progression after it. This biomarker combination is worth validation in larger studies.
    Type of Medium: Online Resource
    ISSN: 2374-4677
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2021
    detail.hit.zdb_id: 2843288-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: npj Breast Cancer, Springer Science and Business Media LLC, Vol. 6, No. 1 ( 2020-05-12)
    Abstract: Stromal tumor-infiltrating lymphocytes (sTILs) are a potential predictive biomarker for immunotherapy response in metastatic triple-negative breast cancer (TNBC). To incorporate sTILs into clinical trials and diagnostics, reliable assessment is essential. In this review, we propose a new concept, namely the implementation of a risk-management framework that enables the use of sTILs as a stratification factor in clinical trials. We present the design of a biomarker risk-mitigation workflow that can be applied to any biomarker incorporation in clinical trials. We demonstrate the implementation of this concept using sTILs as an integral biomarker in a single-center phase II immunotherapy trial for metastatic TNBC (TONIC trial, NCT02499367), using this workflow to mitigate risks of suboptimal inclusion of sTILs in this specific trial. In this review, we demonstrate that a web-based scoring platform can mitigate potential risk factors when including sTILs in clinical trials, and we argue that this framework can be applied for any future biomarker-driven clinical trial setting.
    Type of Medium: Online Resource
    ISSN: 2374-4677
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2020
    detail.hit.zdb_id: 2843288-5
    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