169 results on '"Norma O'Donovan"'
Search Results
2. Preclinical evaluation of Insulin-like growth factor receptor 1 (IGF1R) and Insulin Receptor (IR) as a therapeutic targets in triple negative breast cancer.
- Author
-
Sandra Roche, Patricia Gaule, Deirdre Winrow, Nupur Mukherjee, Fiona O'Neill, Neil T Conlon, Justine Meiller, Denis M Collins, Alexandra Canonici, Mohammed Ibrahim Fawsi, Alejandra Estepa-Fernández, Stephen F Madden, John Crown, Norma O'Donovan, and Alex J Eustace
- Subjects
Medicine ,Science - Abstract
Triple Negative Breast Cancer (TNBC), a subtype of breast cancer, has fewer successful therapeutic therapies than other types of breast cancer. Insulin-like growth factor receptor 1 (IGF1R) and the Insulin receptor (IR) are associated with poor outcomes in TNBC. Targeting IGF1R has failed clinically. We aimed to test if inhibiting both IR/IGF1R was a rationale therapeutic approach to treat TNBC. We showed that despite IGF1R and IR being expressed in TNBC, their expression is not associated with a negative survival outcome. Furthermore, targeting both IR/IGF1R with inhibitors in multiple TNBC cell lines did not inhibit cell growth. Linsitinib, a small molecule inhibitor of both IGF1R and IR, did not block tumour formation and had no effect on tumour growth in vivo. Cumulatively these data suggest that while IGF1R and IR are expressed in TNBC, they are not good therapeutic targets. A potential reason for the limited anti-cancer impact when IR/IGF1R was targeted may be because multiple signalling pathways are altered in TNBC. Therefore, targeting individual signalling pathways may not be sufficient to inhibit cancer growth.
- Published
- 2023
- Full Text
- View/download PDF
3. Identification and clinical impact of potentially actionable somatic oncogenic mutations in solid tumor samples
- Author
-
Sinead Toomey, Aoife Carr, Mateusz Janusz Mezynski, Yasir Elamin, Shereen Rafee, Mattia Cremona, Clare Morgan, Stephen Madden, Khairun I. Abdul-Jalil, Kathy Gately, Angela Farrelly, Elaine W. Kay, Susan Kennedy, Kenneth O’Byrne, Liam Grogan, Oscar Breathnach, Patrick G. Morris, Alexander J. Eustace, Joanna Fay, Robert Cummins, Anthony O’Grady, Roshni Kalachand, Norma O’Donovan, Fergal Kelleher, Aine O’Reilly, Mark Doherty, John Crown, and Bryan T. Hennessy
- Subjects
Medicine - Abstract
Abstract Background An increasing number of anti-cancer therapeutic agents target specific mutant proteins that are expressed by many different tumor types. Successful use of these therapies is dependent on the presence or absence of somatic mutations within the patient’s tumor that can confer clinical efficacy or drug resistance. Methods The aim of our study was to determine the type, frequency, overlap and functional proteomic effects of potentially targetable recurrent somatic hotspot mutations in 47 cancer-related genes in multiple disease sites that could be potential therapeutic targets using currently available agents or agents in clinical development. Results Using MassArray technology, of the 1300 patient tumors analysed 571 (43.9%) had at least one somatic mutation. Mutations were identified in 30 different genes. KRAS (16.5%), PIK3CA (13.6%) and BRAF (3.8%) were the most frequently mutated genes. Prostate (10.8%) had the lowest number of somatic mutations identified, while no mutations were identified in sarcoma. Ocular melanoma (90.6%), endometrial (72.4%) and colorectal (66.4%) tumors had the highest number of mutations. We noted high concordance between mutations in different parts of the tumor (94%) and matched primary and metastatic samples (90%). KRAS and BRAF mutations were mutually exclusive. Mutation co-occurrence involved mainly PIK3CA and PTPN11, and PTPN11 and APC. Reverse Phase Protein Array (RPPA) analysis demonstrated that PI3K and MAPK signalling pathways were more altered in tumors with mutations compared to wild type tumors. Conclusions Hotspot mutational profiling is a sensitive, high-throughput approach for identifying mutations of clinical relevance to molecular based therapeutics for treatment of cancer, and could potentially be of use in identifying novel opportunities for genotype-driven clinical trials.
- Published
- 2020
- Full Text
- View/download PDF
4. Combined targeting EGFR and SRC as a potential novel therapeutic approach for the treatment of triple negative breast cancer
- Author
-
Alexandra Canonici, Alacoque L. Browne, Mohamed F. K. Ibrahim, Kevin P. Fanning, Sandra Roche, Neil T. Conlon, Fiona O’Neill, Justine Meiller, Mattia Cremona, Clare Morgan, Bryan T. Hennessy, Alex J. Eustace, Flavio Solca, Norma O’Donovan, and John Crown
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background: Triple negative breast cancer (TNBC) is an aggressive subtype of breast cancer with limited therapeutic options. Epidermal growth factor receptor (EGFR) has been shown to be over-expressed in TNBC and represents a rational treatment target. Methods: We examined single agent and combination effects for afatinib and dasatinib in TNBC. We then determined IC 50 and combination index values using Calcusyn. Functional analysis of single and combination treatments was performed using reverse phase protein array and cell cycle analysis. Finally, we determined the anticancer effects of the combination in vivo . Results: A total of 14 TNBC cell lines responded to afatinib with IC 50 values ranging from 0.008 to 5.0 µM. Three cell lines, belonging to the basal-like subtype of TNBC, were sensitive to afatinib. The addition of afatinib enhanced response to the five other targeted therapies in HCC1937 and HDQP1 cells. The combination of afatinib with dasatinib caused the greatest growth inhibition in both cell lines. The afatinib/dasatinib combination was synergistic and/or additive in 13/14 TNBC cell lines. Combined afatinib/dasatinib treatment induced G1 cell cycle arrest. Reverse phase protein array results showed the afatinib/dasatinib combination resulted in efficient inhibition of both pERK(T202/T204) and pAkt(S473) signalling in BT20 cells, which was associated with the greatest antiproliferative effects. High baseline levels of pSrc(Y416) and pMAPK(p38) correlated with sensitivity to afatinib, whereas low levels of B-cell lymphoma 2 (Bcl2) and mammalian target of rapamycin (mTOR) correlated with synergistic growth inhibition by combined afatinib and dasatinib treatment. In vivo , the combination treatment inhibited tumour growth in a HCC1806 xenograft model. Conclusions: We demonstrate that afatinib combined with dasatinib has potential clinical activity in TNBC but warrants further preclinical investigation.
- Published
- 2020
- Full Text
- View/download PDF
5. Cleavage of the extracellular domain of junctional adhesion molecule-A is associated with resistance to anti-HER2 therapies in breast cancer settings
- Author
-
Astrid O. Leech, Sri HariKrishna Vellanki, Emily J. Rutherford, Aoife Keogh, Hanne Jahns, Lance Hudson, Norma O’Donovan, Siham Sabri, Bassam Abdulkarim, Katherine M. Sheehan, Elaine W. Kay, Leonie S. Young, Arnold D. K. Hill, Yvonne E. Smith, and Ann M. Hopkins
- Subjects
JAM-A ,Tight junction ,HER2 ,Breast cancer ,Drug resistance ,HER2-targeted therapies ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Junctional adhesion molecule-A (JAM-A) is an adhesion molecule whose overexpression on breast tumor tissue has been associated with aggressive cancer phenotypes, including human epidermal growth factor receptor-2 (HER2)-positive disease. Since JAM-A has been described to regulate HER2 expression in breast cancer cells, we hypothesized that JAM-dependent stabilization of HER2 could participate in resistance to HER2-targeted therapies. Methods Using breast cancer cell line models resistant to anti-HER2 drugs, we investigated JAM-A expression and the effect of JAM-A silencing on biochemical/functional parameters. We also tested whether altered JAM-A expression/processing underpinned differences between drug-sensitive and -resistant cells and acted as a biomarker of patients who developed resistance to HER2-targeted therapies. Results Silencing JAM-A enhanced the anti-proliferative effects of anti-HER2 treatments in trastuzumab- and lapatinib-resistant breast cancer cells and further reduced HER2 protein expression and Akt phosphorylation in drug-treated cells. Increased epidermal growth factor receptor expression observed in drug-resistant models was normalized upon JAM-A silencing. JAM-A was highly expressed in all of a small cohort of HER2-positive patients whose disease recurred following anti-HER2 therapy. High JAM-A expression also correlated with metastatic disease at the time of diagnosis in another patient cohort resistant to trastuzumab therapy. Importantly, cleavage of JAM-A was increased in drug-resistant cell lines in conjunction with increased expression of ADAM-10 and -17 metalloproteases. Pharmacological inhibition or genetic silencing studies suggested a particular role for ADAM-10 in reducing JAM-A cleavage and partially re-sensitizing drug-resistant cells to the anti-proliferative effects of HER2-targeted drugs. Functionally, recombinant cleaved JAM-A enhanced breast cancer cell invasion in vitro and both invasion and proliferation in a semi-in vivo model. Finally, cleaved JAM-A was detectable in the serum of a small cohort of HER2-positive patients and correlated significantly with resistance to HER2-targeted therapy. Conclusions Collectively, our data suggest a novel model whereby increased expression and cleavage of JAM-A drive tumorigenic behavior and act as a biomarker and potential therapeutic target for resistance to HER2-targeted therapies.
- Published
- 2018
- Full Text
- View/download PDF
6. Development of acquired resistance to lapatinib may sensitise HER2-positive breast cancer cells to apoptosis induction by obatoclax and TRAIL
- Author
-
Alex J Eustace, Neil T Conlon, Martina S J McDermott, Brigid C Browne, Patrick O’Leary, Frankie A Holmes, Virginia Espina, Lance A Liotta, Joyce O’Shaughnessy, Clair Gallagher, Lorraine O’Driscoll, Sweta Rani, Stephen F Madden, Neil A O’Brien, Charles Ginther, Dennis Slamon, Naomi Walsh, William M Gallagher, Radoslaw Zagozdzon, William R Watson, Norma O’Donovan, and John Crown
- Subjects
ErbB2 ,MCL-1 ,AKT ,FOXO3a ,cFLIP ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Lapatinib has clinical efficacy in the treatment of trastuzumab-refractory HER2-positive breast cancer. However, a significant proportion of patients develop progressive disease due to acquired resistance to the drug. Induction of apoptotic cell death is a key mechanism of action of lapatinib in HER2-positive breast cancer cells. Methods We examined alterations in regulation of the intrinsic and extrinsic apoptosis pathways in cell line models of acquired lapatinib resistance both in vitro and in patient samples from the NCT01485926 clinical trial, and investigated potential strategies to exploit alterations in apoptosis signalling to overcome lapatinib resistance in HER2-positive breast cancer. Results In this study, we examined two cell lines models of acquired lapatinib resistance (SKBR3-L and HCC1954-L) and showed that lapatinib does not induce apoptosis in these cells. We identified alterations in members of the BCL-2 family of proteins, in particular MCL-1 and BAX, which may play a role in resistance to lapatinib. We tested the therapeutic inhibitor obatoclax, which targets MCL-1. Both SKBR3-L and HCC1954-L cells showed greater sensitivity to obatoclax-induced apoptosis than parental cells. Interestingly, we also found that the development of acquired resistance to lapatinib resulted in acquired sensitivity to TRAIL in SKBR3-L cells. Sensitivity to TRAIL in the SKBR3-L cells was associated with reduced phosphorylation of AKT, increased expression of FOXO3a and decreased expression of c-FLIP. In SKBR3-L cells, TRAIL treatment caused activation of caspase 8, caspase 9 and caspase 3/7. In a second resistant model, HCC1954-L cells, p-AKT levels were not decreased and these cells did not show enhanced sensitivity to TRAIL. Furthermore, combining obatoclax with TRAIL improved response in SKBR3-L cells but not in HCC1954-L cells. Conclusions Our findings highlight the possibility of targeting altered apoptotic signalling to overcome acquired lapatinib resistance, and identify potential novel treatment strategies, with potential biomarkers, for HER2-positive breast cancer that is resistant to HER2 targeted therapies.
- Published
- 2018
- Full Text
- View/download PDF
7. Impact of somatic PI3K pathway and ERBB family mutations on pathological complete response (pCR) in HER2-positive breast cancer patients who received neoadjuvant HER2-targeted therapies
- Author
-
Sinead Toomey, Alexander J. Eustace, Joanna Fay, Katherine M. Sheehan, Aoife Carr, Malgorzata Milewska, Stephen F. Madden, Ausra Teiserskiene, Elaine W. Kay, Norma O’Donovan, William Gallagher, Liam Grogan, Oscar Breathnach, Janice Walshe, Catherine Kelly, Brian Moulton, M. John Kennedy, Guiseppe Gullo, Arnold D. Hill, Colm Power, Deirdre Duke, Niamh Hambly, John Crown, and Bryan T. Hennessy
- Subjects
Breast cancer ,HER2 ,Trastuzumab ,Lapatinib ,PI3K pathway ,Somatic mutations ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background The Cancer Genome Atlas analysis revealed that somatic EGFR, receptor tyrosine-protein kinase erbB-2 (ERBB2), Erb-B2 receptor tyrosine kinase 3 (ERBB3) and Erb-B2 receptor tyrosine kinase 4 (ERBB4) gene mutations (ERBB family mutations) occur alone or co-occur with somatic mutations in the gene encoding the phosphatidylinositol 3-kinase (PI3K) catalytic subunit (PIK3CA) in 19% of human epidermal growth factor receptor 2 (HER2)-positive breast cancers. Because ERBB family mutations can activate the PI3K/AKT pathway and likely have similar canonical signalling effects to PI3K pathway mutations, we investigated their combined impact on response to neoadjuvant HER2-targeted therapies. Methods Baseline tumour biopsies were available from 74 patients with HER2-positive breast cancer who were enrolled in the phase II TCHL neoadjuvant study (ICORG 10-05) assessing TCH (docetaxel, carboplatin, trastuzumab) (n = 38) versus TCL (docetaxel, carboplatin, lapatinib) (n = 10) versus TCHL (docetaxel, carboplatin, trastuzumab, lapatinib) (n = 40), each for six cycles. Activating mutations in PIK3CA and ERBB family genes were identified using mass spectrometry-based genotyping. Phosphatase and tensin homolog (PTEN) expression was assessed by immunohistochemistry. Results PIK3CA and/or ERBB family mutations were detected in 23 (31.1%) tumour samples tested, whereas PTEN expression was low in 31.1% of cases tested. Mutation frequency was similar in each treatment arm (31.3% in TCH arm, 30% in TCL arm and 31.3% in TCHL arm) and was not influenced by oestrogen receptor (ER) status (27.6% in ER-negative patients, 33.3% in ER-positive patients) or progesterone receptor (PR) status (32.6% in PR-negative patients, 29% in PR-positive patients). There was no significant difference in pathological complete response (pCR) rates between 47 patients with wild-type (WT) tumours and 22 patients whose tumours carried mutations (in either PIK3CA or ERBB family genes) (42.5% vs. 54.5%; p = 0.439). Similarly, there was no significant difference in pCR rates between patients with PIK3CA/ERBB family mutated/PTEN-low (i.e., PI3K-activated) tumours and patients without PI3K activation (50% vs. 44%; p = 0.769). However, in the TCHL (but not the TCH) group, the pCR rate was higher for 9 patients with PIK3CA/ERBB family mutated tumours than for 20 patients with PIK3CA/ERBB family WT tumours (77.8% vs. 35%; p = 0.05). Conclusions Our results indicate that patients who receive neoadjuvant TCHL and have PIK3CA/ERBB family mutated tumours may be more likely to have a pCR than patients with WT tumours. Trial registration ClinicalTrials.gov, NCT01485926 . Registered on 2 December 2011.
- Published
- 2017
- Full Text
- View/download PDF
8. Pilot study of bevacizumab in combination with docetaxel and cyclophosphamide as adjuvant treatment for patients with early stage HER-2 negative breast cancer, including analysis of candidate circulating markers of cardiac toxicity: ICORG 08–10 trial
- Author
-
Giuseppe Gullo, Alex J. Eustace, Alexandra Canonici, Denis M. Collins, Michael J. Kennedy, Liam Grogan, Oscar Breathhnach, John McCaffrey, Maccon Keane, Michael J. Martin, Rajnish Gupta, Gregory Leonard, Miriam O’Connor, Paula M. Calvert, Paul Donnellan, Janice Walshe, Enda McDermott, Kathleen Scott, Andres Hernando, Imelda Parker, David W. Murray, Alice C. O’Farrell, Ashwini Maratha, Patrick Dicker, Mairin Rafferty, Verena Murphy, Norma O’Donovan, William M. Gallagher, Bonnie Ky, Dimitrios Tryfonopoulos, Brian Moulton, Annette T. Byrne, and John Crown
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background: Combining bevacizumab and chemotherapy produced superior response rates compared with chemotherapy alone in metastatic breast cancer. As bevacizumab may cause hypertension (HTN) and increase the risk of cardiac failure, we performed a pilot study to evaluate the feasibility and toxicity of a non-anthracycline-containing combination of docetaxel with cyclophosphamide and bevacizumab in early stage breast cancer patients. Methods: Treatment consisted of four 3-weekly cycles of docetaxel and cyclophosphamide (75/600 mg/m 2 ). Bevacizumab was administered 15 mg/kg intravenously on day 1, and then every 3 weeks to a total of 18 cycles of treatment. Serum biomarker concentrations of vascular endothelial growth factor (VEGF), cardiac troponin-I (cTnI), myeloperoxidase (MPO), and placental growth factor (PlGF) were quantified using enzyme-linked immunosorbent assay (ELISA) in 62 patients at baseline and whilst on treatment to determine their utility as biomarkers of cardiotoxicity, indicated by left ventricular ejection fraction (LVEF). Results: A total of 106 patients were accrued in nine sites. Median follow up was 65 months (1–72 months). Seventeen protocol-defined relapse events were observed, accounting for an overall disease-free survival (DFS) rate of 84%. The DFS rates for hormone receptor positive (HR+) and triple-negative (TN) patients were 95% versus 43%, respectively. The median time to relapse was 25 (12–54) months in TN patients versus 38 (22–71) months in HR+ patients. There have been 13 deaths related to breast cancer . The overall survival (OS) rate was 88%. The 5-year OS rate in HR+ versus TN was 95% versus 57%. None of the measured biomarkers predicted the development of cardiotoxicity. Conclusions: We observed a low relapse rate in node-positive, HR+ patients; however, results in TN breast cancer were less encouraging. Given the negative results of three large phase III trials, it is unlikely that this approach will be investigated further. Trial Registration ClinicalTrials.gov Identifier: NCT00911716.
- Published
- 2019
- Full Text
- View/download PDF
9. Identification of potential new treatment response markers and therapeutic targets using a Gaussian process-based method in lapatinib insensitive breast cancer models.
- Author
-
Tapesh Santra, Sandra Roche, Neil Conlon, Norma O'Donovan, John Crown, Robert O'Connor, and Walter Kolch
- Subjects
Medicine ,Science - Abstract
Molecularly targeted therapeutics hold promise of revolutionizing treatments of advanced malignancies. However, a large number of patients do not respond to these treatments. Here, we take a systems biology approach to understand the molecular mechanisms that prevent breast cancer (BC) cells from responding to lapatinib, a dual kinase inhibitor that targets human epidermal growth factor receptor 2 (HER2) and epidermal growth factor receptor (EGFR). To this end, we analysed temporal gene expression profiles of four BC cell lines, two of which respond and the remaining two do not respond to lapatinib. For this analysis, we developed a Gaussian process based algorithm which can accurately find differentially expressed genes by analysing time course gene expression profiles at a fraction of the computational cost of other state-of-the-art algorithms. Our analysis identified 519 potential genes which are characteristic of lapatinib non-responsiveness in the tested cell lines. Data from the Genomics of Drug Sensitivity in Cancer (GDSC) database suggested that the basal expressions 120 of the above genes correlate with the response of BC cells to HER2 and/or EGFR targeted therapies. We selected 27 genes from the larger panel of 519 genes for experimental verification and 16 of these were successfully validated. Further bioinformatics analysis identified vitamin D receptor (VDR) as a potential target of interest for lapatinib non-responsive BC cells. Experimentally, calcitriol, a commonly used reagent for VDR targeted therapy, in combination with lapatinib additively inhibited proliferation in two HER2 positive cell lines, lapatinib insensitive MDA-MB-453 and lapatinib resistant HCC 1954-L cells.
- Published
- 2017
- Full Text
- View/download PDF
10. Supplementary Figure 5 from Effects of HER Family–targeting Tyrosine Kinase Inhibitors on Antibody-dependent Cell-mediated Cytotoxicity in HER2-expressing Breast Cancer
- Author
-
John Crown, Norma O'Donovan, Birgit Bossenmaier, Max Hasmann, Kenneth J. O'Byrne, Bryan T. Hennessy, Lance Liotta, Virginia Espina, Frankie A. Holmes, William M. Gallagher, Darran P. O'Connor, Sinead Toomey, Jo Ballot, Thamir Mahgoub, Anthony M. Davies, Clare Hughes, Kathy A. Gately, Alex J. Eustace, Marion Le Gal, Dalal AlSultan, Nicola Gaynor, Stephen F. Madden, and Denis M. Collins
- Abstract
All ADCC ratios from Figure 3 and trastuzumab/pertuzumab ADCC comparison
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Figure 7 from Effects of HER Family–targeting Tyrosine Kinase Inhibitors on Antibody-dependent Cell-mediated Cytotoxicity in HER2-expressing Breast Cancer
- Author
-
John Crown, Norma O'Donovan, Birgit Bossenmaier, Max Hasmann, Kenneth J. O'Byrne, Bryan T. Hennessy, Lance Liotta, Virginia Espina, Frankie A. Holmes, William M. Gallagher, Darran P. O'Connor, Sinead Toomey, Jo Ballot, Thamir Mahgoub, Anthony M. Davies, Clare Hughes, Kathy A. Gately, Alex J. Eustace, Marion Le Gal, Dalal AlSultan, Nicola Gaynor, Stephen F. Madden, and Denis M. Collins
- Abstract
Densitometry analysis for Figure 6B
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Table 2 from Activated Phosphoinositide 3-Kinase/AKT Signaling Confers Resistance to Trastuzumab but not Lapatinib
- Author
-
Dennis J. Slamon, Norma O'Donovan, John Crown, Michael J. Duffy, Jane Arboleda, Charles Ginther, Yuhua Wang, Lucy Chow, Brigid C. Browne, and Neil A. O'Brien
- Abstract
Supplementary Table 2 from Activated Phosphoinositide 3-Kinase/AKT Signaling Confers Resistance to Trastuzumab but not Lapatinib
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Table 1 from Activated Phosphoinositide 3-Kinase/AKT Signaling Confers Resistance to Trastuzumab but not Lapatinib
- Author
-
Dennis J. Slamon, Norma O'Donovan, John Crown, Michael J. Duffy, Jane Arboleda, Charles Ginther, Yuhua Wang, Lucy Chow, Brigid C. Browne, and Neil A. O'Brien
- Abstract
Supplementary Table 1 from Activated Phosphoinositide 3-Kinase/AKT Signaling Confers Resistance to Trastuzumab but not Lapatinib
- Published
- 2023
- Full Text
- View/download PDF
14. Data from Effects of HER Family–targeting Tyrosine Kinase Inhibitors on Antibody-dependent Cell-mediated Cytotoxicity in HER2-expressing Breast Cancer
- Author
-
John Crown, Norma O'Donovan, Birgit Bossenmaier, Max Hasmann, Kenneth J. O'Byrne, Bryan T. Hennessy, Lance Liotta, Virginia Espina, Frankie A. Holmes, William M. Gallagher, Darran P. O'Connor, Sinead Toomey, Jo Ballot, Thamir Mahgoub, Anthony M. Davies, Clare Hughes, Kathy A. Gately, Alex J. Eustace, Marion Le Gal, Dalal AlSultan, Nicola Gaynor, Stephen F. Madden, and Denis M. Collins
- Abstract
Purpose:Antibody-dependent cell-mediated cytotoxicity (ADCC) is one mechanism of action of the monoclonal antibody (mAb) therapies trastuzumab and pertuzumab. Tyrosine kinase inhibitors (TKIs), like lapatinib, may have added therapeutic value in combination with mAbs through enhanced ADCC activity. Using clinical data, we examined the impact of lapatinib on HER2/EGFR expression levels and natural killer (NK) cell gene signatures. We investigated the ability of three TKIs (lapatinib, afatinib, and neratinib) to alter HER2/immune-related protein levels in preclinical models of HER2-positive (HER2+) and HER2-low breast cancer, and the subsequent effects on trastuzumab/pertuzumab-mediated ADCC.Experimental Design:Preclinical studies (proliferation assays, Western blotting, high content analysis, and flow cytometry) employed HER2+ (SKBR3 and HCC1954) and HER2-low (MCF-7, T47D, CAMA-1, and CAL-51) breast cancer cell lines. NCT00524303 provided reverse phase protein array–determined protein levels of HER2/pHER2/EGFR/pEGFR. RNA-based NK cell gene signatures (CIBERSORT/MCP-counter) post-neoadjuvant anti-HER2 therapy were assessed (NCT00769470/NCT01485926). ADCC assays utilized flow cytometry–based protocols.Results:Lapatinib significantly increased membrane HER2 levels, while afatinib and neratinib significantly decreased levels in all preclinical models. Single-agent lapatinib increased HER2 or EGFR levels in 10 of 11 (91%) tumor samples. NK cell signatures increased posttherapy (P = 0.03) and associated with trastuzumab response (P = 0.01). TKI treatment altered mAb-induced NK cell–mediated ADCC in vitro, but it did not consistently correlate with HER2 expression in HER2+ or HER2-low models. The ADCC response to trastuzumab and pertuzumab combined did not exceed either mAb alone.Conclusions:TKIs differentially alter tumor cell phenotype which can impact NK cell–mediated response to coadministered antibody therapies. mAb-induced ADCC response is relevant when rationalizing combinations for clinical investigation.
- Published
- 2023
- Full Text
- View/download PDF
15. Supplementary Figure 3 from Effects of HER Family–targeting Tyrosine Kinase Inhibitors on Antibody-dependent Cell-mediated Cytotoxicity in HER2-expressing Breast Cancer
- Author
-
John Crown, Norma O'Donovan, Birgit Bossenmaier, Max Hasmann, Kenneth J. O'Byrne, Bryan T. Hennessy, Lance Liotta, Virginia Espina, Frankie A. Holmes, William M. Gallagher, Darran P. O'Connor, Sinead Toomey, Jo Ballot, Thamir Mahgoub, Anthony M. Davies, Clare Hughes, Kathy A. Gately, Alex J. Eustace, Marion Le Gal, Dalal AlSultan, Nicola Gaynor, Stephen F. Madden, and Denis M. Collins
- Abstract
Densitometry for Figure 1D
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary Figure 8 from Effects of HER Family–targeting Tyrosine Kinase Inhibitors on Antibody-dependent Cell-mediated Cytotoxicity in HER2-expressing Breast Cancer
- Author
-
John Crown, Norma O'Donovan, Birgit Bossenmaier, Max Hasmann, Kenneth J. O'Byrne, Bryan T. Hennessy, Lance Liotta, Virginia Espina, Frankie A. Holmes, William M. Gallagher, Darran P. O'Connor, Sinead Toomey, Jo Ballot, Thamir Mahgoub, Anthony M. Davies, Clare Hughes, Kathy A. Gately, Alex J. Eustace, Marion Le Gal, Dalal AlSultan, Nicola Gaynor, Stephen F. Madden, and Denis M. Collins
- Abstract
Densitometry analysis for Figure 6D/E
- Published
- 2023
- Full Text
- View/download PDF
17. Supplementary Tables 1-4 from Effects of HER Family–targeting Tyrosine Kinase Inhibitors on Antibody-dependent Cell-mediated Cytotoxicity in HER2-expressing Breast Cancer
- Author
-
John Crown, Norma O'Donovan, Birgit Bossenmaier, Max Hasmann, Kenneth J. O'Byrne, Bryan T. Hennessy, Lance Liotta, Virginia Espina, Frankie A. Holmes, William M. Gallagher, Darran P. O'Connor, Sinead Toomey, Jo Ballot, Thamir Mahgoub, Anthony M. Davies, Clare Hughes, Kathy A. Gately, Alex J. Eustace, Marion Le Gal, Dalal AlSultan, Nicola Gaynor, Stephen F. Madden, and Denis M. Collins
- Abstract
Flow cytometry MFI and % positive cells data for fluorescently labelled trastuzumab and pertuzumab in SKBR3, HCC1954, T47D and MCF-7 treated with TKIs
- Published
- 2023
- Full Text
- View/download PDF
18. Supplementary Data Figure 1 from Effects of HER Family–targeting Tyrosine Kinase Inhibitors on Antibody-dependent Cell-mediated Cytotoxicity in HER2-expressing Breast Cancer
- Author
-
John Crown, Norma O'Donovan, Birgit Bossenmaier, Max Hasmann, Kenneth J. O'Byrne, Bryan T. Hennessy, Lance Liotta, Virginia Espina, Frankie A. Holmes, William M. Gallagher, Darran P. O'Connor, Sinead Toomey, Jo Ballot, Thamir Mahgoub, Anthony M. Davies, Clare Hughes, Kathy A. Gately, Alex J. Eustace, Marion Le Gal, Dalal AlSultan, Nicola Gaynor, Stephen F. Madden, and Denis M. Collins
- Abstract
Rituximab ADCC negative control
- Published
- 2023
- Full Text
- View/download PDF
19. Supplementary Figure 2 from Effects of HER Family–targeting Tyrosine Kinase Inhibitors on Antibody-dependent Cell-mediated Cytotoxicity in HER2-expressing Breast Cancer
- Author
-
John Crown, Norma O'Donovan, Birgit Bossenmaier, Max Hasmann, Kenneth J. O'Byrne, Bryan T. Hennessy, Lance Liotta, Virginia Espina, Frankie A. Holmes, William M. Gallagher, Darran P. O'Connor, Sinead Toomey, Jo Ballot, Thamir Mahgoub, Anthony M. Davies, Clare Hughes, Kathy A. Gately, Alex J. Eustace, Marion Le Gal, Dalal AlSultan, Nicola Gaynor, Stephen F. Madden, and Denis M. Collins
- Abstract
Flow cytometry control scatter plots for trastuzumab-488 and pertuzumab-550
- Published
- 2023
- Full Text
- View/download PDF
20. Supplementary Figure 6 from Effects of HER Family–targeting Tyrosine Kinase Inhibitors on Antibody-dependent Cell-mediated Cytotoxicity in HER2-expressing Breast Cancer
- Author
-
John Crown, Norma O'Donovan, Birgit Bossenmaier, Max Hasmann, Kenneth J. O'Byrne, Bryan T. Hennessy, Lance Liotta, Virginia Espina, Frankie A. Holmes, William M. Gallagher, Darran P. O'Connor, Sinead Toomey, Jo Ballot, Thamir Mahgoub, Anthony M. Davies, Clare Hughes, Kathy A. Gately, Alex J. Eustace, Marion Le Gal, Dalal AlSultan, Nicola Gaynor, Stephen F. Madden, and Denis M. Collins
- Abstract
MFI comparison at T0 and T6 for T47D/MCF-7
- Published
- 2023
- Full Text
- View/download PDF
21. Data from Activated Phosphoinositide 3-Kinase/AKT Signaling Confers Resistance to Trastuzumab but not Lapatinib
- Author
-
Dennis J. Slamon, Norma O'Donovan, John Crown, Michael J. Duffy, Jane Arboleda, Charles Ginther, Yuhua Wang, Lucy Chow, Brigid C. Browne, and Neil A. O'Brien
- Abstract
Trastuzumab and lapatinib provide clinical benefit to women with human epidermal growth factor receptor 2 (HER)–positive breast cancer. However, not all patients whose tumors contain the HER2 alteration respond. Consequently, there is an urgent need to identify new predictive factors for these agents. The aim of this study was to investigate the role of receptor tyrosine kinase signaling and phosphoinositide 3-kinase (PI3K)/AKT pathway activation in conferring resistance to trastuzumab and lapatinib. To address this question, we evaluated response to trastuzumab and lapatinib in a panel of 18 HER2-amplified cell lines, using both two- and three-dimensional culture. The SUM-225, HCC-1419, HCC-1954, UACC-893, HCC-1569, UACC-732, JIMT-1, and MDA-453 cell lines were found to be innately resistant to trastuzumab, whereas the MDA-361, MDA-453, HCC-1569, UACC-732, JIMT-1, HCC-202, and UACC-893 cells are innately lapatinib resistant. Lapatinib was active in de novo (SUM-225, HCC-1419, and HCC-1954) and in a BT-474 cell line with acquired resistance to trastuzumab. In these cells, trastuzumab had little effect on AKT phosphorylation, whereas lapatinib retained activity through the dephosphorylation of AKT. Increased phosphorylation of HER2, epidermal growth factor receptor, HER3, and insulin-like growth factor IR correlated with response to lapatinib but not trastuzumab. Loss of PTEN or the presence of activating mutations in PI3K marked resistance to trastuzumab, but lapatinib response was independent of these factors. Thus, increased activation of the PI3K/AKT pathway correlates with resistance to trastuzumab, which can be overcome by lapatinib. In conclusion, pharmacologic targeting of the PI3K/AKT pathway may provide benefit to HER2-positive breast cancer patients who are resistant to trastuzumab therapy. Mol Cancer Ther; 9(6); 1489–502. ©2010 AACR.
- Published
- 2023
- Full Text
- View/download PDF
22. Supplementary Figure 4 from Effects of HER Family–targeting Tyrosine Kinase Inhibitors on Antibody-dependent Cell-mediated Cytotoxicity in HER2-expressing Breast Cancer
- Author
-
John Crown, Norma O'Donovan, Birgit Bossenmaier, Max Hasmann, Kenneth J. O'Byrne, Bryan T. Hennessy, Lance Liotta, Virginia Espina, Frankie A. Holmes, William M. Gallagher, Darran P. O'Connor, Sinead Toomey, Jo Ballot, Thamir Mahgoub, Anthony M. Davies, Clare Hughes, Kathy A. Gately, Alex J. Eustace, Marion Le Gal, Dalal AlSultan, Nicola Gaynor, Stephen F. Madden, and Denis M. Collins
- Abstract
Densitometry for Figure 2B
- Published
- 2023
- Full Text
- View/download PDF
23. Suppl Fig. S3 from Neuromedin U: A Candidate Biomarker and Therapeutic Target to Predict and Overcome Resistance to HER-Tyrosine Kinase Inhibitors
- Author
-
Lorraine O'Driscoll, Annette T. Byrne, Martina Gogarty, John Crown, Norma O'Donovan, Brigid C. Browne, Martina S. McDermott, Stephen Madden, Susan Breslin, Serena Germano, Liam Shiels, Claire Corcoran, and Sweta Rani
- Abstract
Suppl Fig. S3
- Published
- 2023
- Full Text
- View/download PDF
24. Suppl Table 2 from Neuromedin U: A Candidate Biomarker and Therapeutic Target to Predict and Overcome Resistance to HER-Tyrosine Kinase Inhibitors
- Author
-
Lorraine O'Driscoll, Annette T. Byrne, Martina Gogarty, John Crown, Norma O'Donovan, Brigid C. Browne, Martina S. McDermott, Stephen Madden, Susan Breslin, Serena Germano, Liam Shiels, Claire Corcoran, and Sweta Rani
- Abstract
Suppl Table 2
- Published
- 2023
- Full Text
- View/download PDF
25. Suppl Material & Suppl. Table S1 from Neuromedin U: A Candidate Biomarker and Therapeutic Target to Predict and Overcome Resistance to HER-Tyrosine Kinase Inhibitors
- Author
-
Lorraine O'Driscoll, Annette T. Byrne, Martina Gogarty, John Crown, Norma O'Donovan, Brigid C. Browne, Martina S. McDermott, Stephen Madden, Susan Breslin, Serena Germano, Liam Shiels, Claire Corcoran, and Sweta Rani
- Abstract
Suppl Material & Suppl. Table S1
- Published
- 2023
- Full Text
- View/download PDF
26. Data from Neuromedin U: A Candidate Biomarker and Therapeutic Target to Predict and Overcome Resistance to HER-Tyrosine Kinase Inhibitors
- Author
-
Lorraine O'Driscoll, Annette T. Byrne, Martina Gogarty, John Crown, Norma O'Donovan, Brigid C. Browne, Martina S. McDermott, Stephen Madden, Susan Breslin, Serena Germano, Liam Shiels, Claire Corcoran, and Sweta Rani
- Abstract
Intrinsic and acquired resistance to HER-targeting drugs occurs in a significant proportion of HER2-overexpressing breast cancers. Thus, there remains a need to identify predictive biomarkers that could improve patient selection and circumvent these types of drug resistance. Here, we report the identification of neuromedin U (NmU) as an extracellular biomarker in cells resistant to HER-targeted drugs. NmU overexpression occurred in cells with acquired or innate resistance to lapatinib, trastuzumab, neratinib, and afatinib, all of which displayed a similar trend upon short-term exposure, suggesting NmU induction may be an early response. An analysis of 3,489 cases of breast cancer showed NmU to be associated with poor patient outcome, particularly those with HER2-overexpressing tumors independent of established prognostic indicators. Ectopic overexpression of NmU in drug-sensitive cells conferred resistance to all HER-targeting drugs, whereas RNAi-mediated attenuation sensitized cells exhibiting acquired or innate drug resistance. Mechanistic investigations suggested that NmU acted through HSP27 as partner protein to stabilize HER2 protein levels. We also obtained evidence of functional NmU receptors on HER2-overexpressing cells, with the addition of exogenous NmU eliciting an elevation in HER2 and EGFR expression along with drug resistance. Finally, we found that NmU seemed to function in cell motility, invasion, and anoikis resistance. In vivo studies revealed that NmU attenuation impaired tumor growth and metastasis. Taken together, our results defined NmU as a candidate drug response biomarker for HER2-overexpressing cancers and as a candidate therapeutic target to limit metastatic progression and improve the efficacy of HER-targeted drugs. Cancer Res; 74(14); 3821–33. ©2014 AACR.
- Published
- 2023
- Full Text
- View/download PDF
27. A Systems Biology Approach to Investigate Kinase Signal Transduction Networks That Are Involved in Triple Negative Breast Cancer Resistance to Cisplatin
- Author
-
Nupur Mukherjee, Alacoque Browne, Laura Ivers, Tapesh Santra, Mattia Cremona, Bryan T. Hennessy, Norma O’Donovan, John Crown, Walter Kolch, Dirk Fey, and Alex J. Eustace
- Subjects
Medicine (miscellaneous) ,triple negative breast cancer ,reverse-phase protein array ,system biology ,BMRA analysis ,cisplatin resistance ,P53 mutation ,PI3K/AKT pathway - Abstract
Triple negative breast cancer (TNBC) remains a therapeutic challenge due to the lack of targetable genetic alterations and the frequent development of resistance to the standard cisplatin-based chemotherapies. Here, we have taken a systems biology approach to investigate kinase signal transduction networks that are involved in TNBC resistance to cisplatin. Treating a panel of cisplatin-sensitive and cisplatin-resistant TNBC cell lines with a panel of kinase inhibitors allowed us to reconstruct two kinase signalling networks that characterise sensitive and resistant cells. The analysis of these networks suggested that the activation of the PI3K/AKT signalling pathway is critical for cisplatin resistance. Experimental validation of the computational model predictions confirmed that TNBC cell lines with activated PI3K/AKT signalling are sensitive to combinations of cisplatin and PI3K/AKT pathway inhibitors. Thus, our results reveal a new therapeutic approach that is based on identifying targeted therapies that synergise with conventional chemotherapies.
- Published
- 2022
- Full Text
- View/download PDF
28. Inhibition of CDK8/19 Mediator kinase potentiates HER2-targeting drugs and bypasses resistance to these agents in vitro and in vivo
- Author
-
Xiaokai Ding, Amanda C. Sharko, Martina S. J. McDermott, Gary P. Schools, Alexander Chumanevich, Hao Ji, Jing Li, Li Zhang, Zachary T. Mack, Vitali Sikirzhytski, Michael Shtutman, Laura Ivers, Norma O’Donovan, John Crown, Balázs Győrffy, Mengqian Chen, Igor B. Roninson, and Eugenia V. Broude
- Subjects
Multidisciplinary ,Receptor, ErbB-2 ,Antineoplastic Agents ,Breast Neoplasms ,Lapatinib ,Trastuzumab ,Cyclin-Dependent Kinase 8 ,Xenograft Model Antitumor Assays ,Cyclin-Dependent Kinases ,Mice ,Phosphatidylinositol 3-Kinases ,Drug Resistance, Neoplasm ,Cell Line, Tumor ,Animals ,Humans ,Female ,Protein Kinase Inhibitors - Abstract
Breast cancers (BrCas) that overexpress oncogenic tyrosine kinase receptor HER2 are treated with HER2-targeting antibodies (such as trastuzumab) or small-molecule kinase inhibitors (such as lapatinib). However, most patients with metastatic HER2+BrCa have intrinsic resistance and nearly all eventually become resistant to HER2-targeting therapy. Resistance to HER2-targeting drugs frequently involves transcriptional reprogramming associated with constitutive activation of different signaling pathways. We have investigated the role of CDK8/19 Mediator kinase, a regulator of transcriptional reprogramming, in the response of HER2+BrCa to HER2-targeting drugs. CDK8 was in the top 1% of all genes ranked by correlation with shorter relapse-free survival among treated HER2+BrCa patients. Selective CDK8/19 inhibitors (senexin B and SNX631) showed synergistic interactions with lapatinib and trastuzumab in a panel of HER2+BrCa cell lines, overcoming and preventing resistance to HER2-targeting drugs. The synergistic effects were mediated in part through the PI3K/AKT/mTOR pathway and reduced by PI3K inhibition. Combination of HER2- and CDK8/19-targeting agents inhibited STAT1 and STAT3 phosphorylation at S727 and up-regulated tumor suppressor BTG2. The growth of xenograft tumors formed by lapatinib-sensitive or -resistant HER2+breast cancer cells was partially inhibited by SNX631 alone and strongly suppressed by the combination of SNX631 and lapatinib, overcoming lapatinib resistance. These effects were associated with decreased tumor cell proliferation and altered recruitment of stromal components to the xenograft tumors. These results suggest potential clinical benefit of combining HER2- and CDK8/19-targeting drugs in the treatment of metastatic HER2+BrCa.
- Published
- 2022
- Full Text
- View/download PDF
29. The role of infiltrating lymphocytes in the neo-adjuvant treatment of women with HER2-positive breast cancer
- Author
-
Stephen F. Madden, Colm Power, Janice M. Walshe, Simon J Furney, John Crown, Alex J Eustace, Sinead Toomey, A Hill, William M. Gallagher, Katherine M. Sheehan, Ailis Fagan, Oscar S. Breathnach, Bryan T. Hennessy, Deirdre Duke, Liam Grogan, Bruce Moran, Patrick G. Morris, Norma O'Donovan, Ausra Teiserskiene, Joanna Fay, Elaine W. Kay, Denis M. Collins, and K. Egan
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Receptor, ErbB-2 ,Lymphocyte ,medicine.medical_treatment ,Breast Neoplasms ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Immune system ,Preclinical Study ,Lymphocytes, Tumor-Infiltrating ,Internal medicine ,Tumour infiltrating lymphocytes ,Antineoplastic Combined Chemotherapy Protocols ,Medicine ,Humans ,Lymphocytes ,HER2-positive breast cancer ,Pathological ,Neoadjuvant therapy ,Chemotherapy ,business.industry ,T-cells ,medicine.disease ,Neo-adjuvant treatment ,Prognosis ,Neoadjuvant Therapy ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cohort ,Immunohistochemistry ,Female ,business - Abstract
Background Pre-treatment tumour-associated lymphocytes (TILs) and stromal lymphocytes (SLs) are independent predictive markers of future pathological complete response (pCR) in HER2-positive breast cancer. Whilst studies have correlated baseline lymphocyte levels with subsequent pCR, few have studied the impact of neoadjuvant therapy on the immune environment. Methods We performed TIL analysis and T-cell analysis by IHC on the pretreatment and ‘On-treatment’ samples from patients recruited on the Phase-II TCHL (NCT01485926) clinical trial. Data were analysed using the Wilcoxon signed-rank test and the Spearman rank correlation. Results In our sample cohort (n = 66), patients who achieved a pCR at surgery, post-chemotherapy, had significantly higher counts of TILs (p = 0.05) but not SLs (p = 0.08) in their pre-treatment tumour samples. Patients who achieved a subsequent pCR after completing neo-adjuvant chemotherapy had significantly higher SLs (p = 9.09 × 10–3) but not TILs (p = 0.1) in their ‘On-treatment’ tumour biopsies. In a small cohort of samples (n = 16), infiltrating lymphocyte counts increased after 1 cycle of neo-adjuvant chemotherapy only in those tumours of patients who did not achieve a subsequent pCR. Finally, reduced CD3 + (p = 0.04, rho = 0.60) and CD4 + (p = 0.01, rho = 0.72) T-cell counts in 'On-treatment' biopsies were associated with decreased residual tumour content post-1 cycle of treatment; the latter being significantly associated with increased likelihood of subsequent pCR (p Conclusions The immune system may be ‘primed’ prior to neoadjuvant treatment in those patients who subsequently achieve a pCR. In those patients who achieve a pCR, their immune response may return to baseline after only 1 cycle of treatment. However, in those who did not achieve a pCR, neo-adjuvant treatment may stimulate lymphocyte influx into the tumour.
- Published
- 2021
30. Effects of HER Family–targeting Tyrosine Kinase Inhibitors on Antibody-dependent Cell-mediated Cytotoxicity in HER2-expressing Breast Cancer
- Author
-
Thamir Mahgoub, Kenneth J. O'Byrne, John Crown, Clare Hughes, Frankie A. Holmes, Virginia Espina, Anthony Davies, Marion Le Gal, Alex J Eustace, Lance A. Liotta, Kathy Gately, Norma O'Donovan, Darran P. O'Connor, Max Hasmann, Denis M. Collins, Sinead Toomey, Dalal Alsultan, Jo Ballot, Birgit Bossenmaier, N. Gaynor, Stephen F. Madden, Bryan T. Hennessy, and William M. Gallagher
- Subjects
Adult ,0301 basic medicine ,Cancer Research ,Adolescent ,Receptor, ErbB-2 ,Afatinib ,Breast Neoplasms ,Antibodies, Monoclonal, Humanized ,Lapatinib ,Article ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Trastuzumab ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,RNA-Seq ,skin and connective tissue diseases ,Protein Kinase Inhibitors ,neoplasms ,Aged ,Antibody-dependent cell-mediated cytotoxicity ,business.industry ,Antibody-Dependent Cell Cytotoxicity ,Middle Aged ,Neoadjuvant Therapy ,Gene Expression Regulation, Neoplastic ,Killer Cells, Natural ,030104 developmental biology ,Oncology ,SKBR3 ,030220 oncology & carcinogenesis ,Neratinib ,MCF-7 Cells ,Cancer research ,Female ,Pertuzumab ,business ,Tyrosine kinase ,medicine.drug - Abstract
Purpose: Antibody-dependent cell-mediated cytotoxicity (ADCC) is one mechanism of action of the monoclonal antibody (mAb) therapies trastuzumab and pertuzumab. Tyrosine kinase inhibitors (TKIs), like lapatinib, may have added therapeutic value in combination with mAbs through enhanced ADCC activity. Using clinical data, we examined the impact of lapatinib on HER2/EGFR expression levels and natural killer (NK) cell gene signatures. We investigated the ability of three TKIs (lapatinib, afatinib, and neratinib) to alter HER2/immune-related protein levels in preclinical models of HER2-positive (HER2+) and HER2-low breast cancer, and the subsequent effects on trastuzumab/pertuzumab-mediated ADCC. Experimental Design: Preclinical studies (proliferation assays, Western blotting, high content analysis, and flow cytometry) employed HER2+ (SKBR3 and HCC1954) and HER2-low (MCF-7, T47D, CAMA-1, and CAL-51) breast cancer cell lines. NCT00524303 provided reverse phase protein array–determined protein levels of HER2/pHER2/EGFR/pEGFR. RNA-based NK cell gene signatures (CIBERSORT/MCP-counter) post-neoadjuvant anti-HER2 therapy were assessed (NCT00769470/NCT01485926). ADCC assays utilized flow cytometry–based protocols. Results: Lapatinib significantly increased membrane HER2 levels, while afatinib and neratinib significantly decreased levels in all preclinical models. Single-agent lapatinib increased HER2 or EGFR levels in 10 of 11 (91%) tumor samples. NK cell signatures increased posttherapy (P = 0.03) and associated with trastuzumab response (P = 0.01). TKI treatment altered mAb-induced NK cell–mediated ADCC in vitro, but it did not consistently correlate with HER2 expression in HER2+ or HER2-low models. The ADCC response to trastuzumab and pertuzumab combined did not exceed either mAb alone. Conclusions: TKIs differentially alter tumor cell phenotype which can impact NK cell–mediated response to coadministered antibody therapies. mAb-induced ADCC response is relevant when rationalizing combinations for clinical investigation.
- Published
- 2021
- Full Text
- View/download PDF
31. Identification and clinical impact of potentially actionable somatic oncogenic mutations in solid tumor samples
- Author
-
Oscar S. Breathnach, Kathy Gately, Robert Cummins, Mattia Cremona, Elaine W. Kay, A. O'Reilly, Khairun I. Abdul-Jalil, Alex J Eustace, Mateusz Janusz Mezynski, Kenneth J. O'Byrne, Patrick G. Morris, Norma O'Donovan, Susan Kennedy, Joanna Fay, Stephen F. Madden, Fergal C. Kelleher, Liam Grogan, John Crown, Anthony O'Grady, Clare Morgan, Mark A. Doherty, Roshni Kalachand, Aoife Carr, Bryan T. Hennessy, Shereen Rafee, Sinead Toomey, Angela M. Farrelly, and Yasir Y. Elamin
- Subjects
Male ,Proteomics ,Proto-Oncogene Proteins B-raf ,0301 basic medicine ,Class I Phosphatidylinositol 3-Kinases ,Somatic cell ,lcsh:Medicine ,Antineoplastic Agents ,Biology ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Proto-Oncogene Proteins p21(ras) ,03 medical and health sciences ,0302 clinical medicine ,Germline mutation ,medicine ,Humans ,Gene ,Research ,lcsh:R ,Wild type ,Reverse phase protein lysate microarray ,Oncogenes ,General Medicine ,medicine.disease ,PTPN11 ,030104 developmental biology ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Sarcoma ,KRAS ,Colorectal Neoplasms ,Signal Transduction - Abstract
Background An increasing number of anti-cancer therapeutic agents target specific mutant proteins that are expressed by many different tumor types. Successful use of these therapies is dependent on the presence or absence of somatic mutations within the patient’s tumor that can confer clinical efficacy or drug resistance. Methods The aim of our study was to determine the type, frequency, overlap and functional proteomic effects of potentially targetable recurrent somatic hotspot mutations in 47 cancer-related genes in multiple disease sites that could be potential therapeutic targets using currently available agents or agents in clinical development. Results Using MassArray technology, of the 1300 patient tumors analysed 571 (43.9%) had at least one somatic mutation. Mutations were identified in 30 different genes. KRAS (16.5%), PIK3CA (13.6%) and BRAF (3.8%) were the most frequently mutated genes. Prostate (10.8%) had the lowest number of somatic mutations identified, while no mutations were identified in sarcoma. Ocular melanoma (90.6%), endometrial (72.4%) and colorectal (66.4%) tumors had the highest number of mutations. We noted high concordance between mutations in different parts of the tumor (94%) and matched primary and metastatic samples (90%). KRAS and BRAF mutations were mutually exclusive. Mutation co-occurrence involved mainly PIK3CA and PTPN11, and PTPN11 and APC. Reverse Phase Protein Array (RPPA) analysis demonstrated that PI3K and MAPK signalling pathways were more altered in tumors with mutations compared to wild type tumors. Conclusions Hotspot mutational profiling is a sensitive, high-throughput approach for identifying mutations of clinical relevance to molecular based therapeutics for treatment of cancer, and could potentially be of use in identifying novel opportunities for genotype-driven clinical trials.
- Published
- 2020
- Full Text
- View/download PDF
32. Cleavage of the extracellular domain of junctional adhesion molecule-A is associated with resistance to anti-HER2 therapies in breast cancer settings
- Author
-
Aoife Keogh, Bassam Abdulkarim, Arnold D.K. Hill, Hanne Jahns, Katherine M. Sheehan, Leonie S. Young, Emily J. Rutherford, Siham Sabri, Astrid O. Leech, Elaine W. Kay, Yvonne E. Smith, Lance Hudson, Norma O'Donovan, Sri HariKrishna Vellanki, and Ann M. Hopkins
- Subjects
0301 basic medicine ,Receptor, ErbB-2 ,Neoplasm invasiveness ,Chick Embryo ,Chorioallantoic Membrane ,0302 clinical medicine ,Antineoplastic Agents, Immunological ,Breast cancer ,Trastuzumab ,ErbB-2 receptor ,Cell Movement ,Epidermal growth factor receptor ,RNA, Small Interfering ,skin and connective tissue diseases ,JAM-A ,biology ,Cell adhesion molecule ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Recombinant Proteins ,humanities ,3. Good health ,030220 oncology & carcinogenesis ,Immunological antineoplastic agents ,Female ,medicine.drug ,Junctional Adhesion Molecule A ,Research Article ,Breast Neoplasms ,Receptors, Cell Surface ,Lapatinib ,lcsh:RC254-282 ,03 medical and health sciences ,Herceptin ,Cell Line, Tumor ,HER2 ,medicine ,Biomarkers, Tumor ,Gene silencing ,Animals ,Humans ,Neoplasm Invasiveness ,HER2-targeted therapies ,Tight junction ,business.industry ,fungi ,Tumor biomarkers ,medicine.disease ,030104 developmental biology ,Cell culture ,Drug Resistance, Neoplasm ,Drug resistance ,Cancer research ,biology.protein ,business ,Cell Adhesion Molecules - Abstract
Background Junctional adhesion molecule-A (JAM-A) is an adhesion molecule whose overexpression on breast tumor tissue has been associated with aggressive cancer phenotypes, including human epidermal growth factor receptor-2 (HER2)-positive disease. Since JAM-A has been described to regulate HER2 expression in breast cancer cells, we hypothesized that JAM-dependent stabilization of HER2 could participate in resistance to HER2-targeted therapies. Methods Using breast cancer cell line models resistant to anti-HER2 drugs, we investigated JAM-A expression and the effect of JAM-A silencing on biochemical/functional parameters. We also tested whether altered JAM-A expression/processing underpinned differences between drug-sensitive and -resistant cells and acted as a biomarker of patients who developed resistance to HER2-targeted therapies. Results Silencing JAM-A enhanced the anti-proliferative effects of anti-HER2 treatments in trastuzumab- and lapatinib-resistant breast cancer cells and further reduced HER2 protein expression and Akt phosphorylation in drug-treated cells. Increased epidermal growth factor receptor expression observed in drug-resistant models was normalized upon JAM-A silencing. JAM-A was highly expressed in all of a small cohort of HER2-positive patients whose disease recurred following anti-HER2 therapy. High JAM-A expression also correlated with metastatic disease at the time of diagnosis in another patient cohort resistant to trastuzumab therapy. Importantly, cleavage of JAM-A was increased in drug-resistant cell lines in conjunction with increased expression of ADAM-10 and -17 metalloproteases. Pharmacological inhibition or genetic silencing studies suggested a particular role for ADAM-10 in reducing JAM-A cleavage and partially re-sensitizing drug-resistant cells to the anti-proliferative effects of HER2-targeted drugs. Functionally, recombinant cleaved JAM-A enhanced breast cancer cell invasion in vitro and both invasion and proliferation in a semi-in vivo model. Finally, cleaved JAM-A was detectable in the serum of a small cohort of HER2-positive patients and correlated significantly with resistance to HER2-targeted therapy. Conclusions Collectively, our data suggest a novel model whereby increased expression and cleavage of JAM-A drive tumorigenic behavior and act as a biomarker and potential therapeutic target for resistance to HER2-targeted therapies. Electronic supplementary material The online version of this article (10.1186/s13058-018-1064-1) contains supplementary material, which is available to authorized users.
- Published
- 2018
- Full Text
- View/download PDF
33. Development of acquired resistance to lapatinib may sensitise HER2-positive breast cancer cells to apoptosis induction by obatoclax and TRAIL
- Author
-
Neil T Conlon, Patrick O’Leary, Virginia Espina, William Watson, John Crown, Frankie A. Holmes, Sweta Rani, William M. Gallagher, Lance A. Liotta, Radoslaw Zagozdzon, Norma O'Donovan, Naomi Walsh, Alex J Eustace, Stephen F. Madden, Charles Ginther, Brigid C. Browne, Martina McDermott, Joyce O'Shaughnessy, Dennis J. Slamon, Neil A. O'Brien, Lorraine O'Driscoll, and Clair Gallagher
- Subjects
0301 basic medicine ,Cancer Research ,Drug Resistance ,CASP8 and FADD-Like Apoptosis Regulating Protein ,Gene Expression ,Apoptosis ,Drug resistance ,TNF-Related Apoptosis-Inducing Ligand ,chemistry.chemical_compound ,0302 clinical medicine ,ErbB2 ,2.1 Biological and endogenous factors ,Phosphorylation ,Aetiology ,skin and connective tissue diseases ,Cancer ,Tumor ,Forkhead Box Protein O3 ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,3. Good health ,Oncology ,Proto-Oncogene Proteins c-bcl-2 ,5.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Public Health and Health Services ,Female ,Development of treatments and therapeutic interventions ,Research Article ,medicine.drug ,Oncology and Carcinogenesis ,Caspase 3 ,Breast Neoplasms ,Antineoplastic Agents ,Lapatinib ,Caspase 8 ,lcsh:RC254-282 ,Cell Line ,03 medical and health sciences ,Breast cancer ,Cell Line, Tumor ,Breast Cancer ,Genetics ,medicine ,Humans ,Oncology & Carcinogenesis ,FOXO3a ,Protein kinase B ,erbB-2 ,business.industry ,AKT ,cFLIP ,Genes, erbB-2 ,medicine.disease ,030104 developmental biology ,chemistry ,Genes ,Drug Resistance, Neoplasm ,Cancer research ,Neoplasm ,MCL-1 ,business ,Obatoclax - Abstract
Background Lapatinib has clinical efficacy in the treatment of trastuzumab-refractory HER2-positive breast cancer. However, a significant proportion of patients develop progressive disease due to acquired resistance to the drug. Induction of apoptotic cell death is a key mechanism of action of lapatinib in HER2-positive breast cancer cells. Methods We examined alterations in regulation of the intrinsic and extrinsic apoptosis pathways in cell line models of acquired lapatinib resistance both in vitro and in patient samples from the NCT01485926 clinical trial, and investigated potential strategies to exploit alterations in apoptosis signalling to overcome lapatinib resistance in HER2-positive breast cancer. Results In this study, we examined two cell lines models of acquired lapatinib resistance (SKBR3-L and HCC1954-L) and showed that lapatinib does not induce apoptosis in these cells. We identified alterations in members of the BCL-2 family of proteins, in particular MCL-1 and BAX, which may play a role in resistance to lapatinib. We tested the therapeutic inhibitor obatoclax, which targets MCL-1. Both SKBR3-L and HCC1954-L cells showed greater sensitivity to obatoclax-induced apoptosis than parental cells. Interestingly, we also found that the development of acquired resistance to lapatinib resulted in acquired sensitivity to TRAIL in SKBR3-L cells. Sensitivity to TRAIL in the SKBR3-L cells was associated with reduced phosphorylation of AKT, increased expression of FOXO3a and decreased expression of c-FLIP. In SKBR3-L cells, TRAIL treatment caused activation of caspase 8, caspase 9 and caspase 3/7. In a second resistant model, HCC1954-L cells, p-AKT levels were not decreased and these cells did not show enhanced sensitivity to TRAIL. Furthermore, combining obatoclax with TRAIL improved response in SKBR3-L cells but not in HCC1954-L cells. Conclusions Our findings highlight the possibility of targeting altered apoptotic signalling to overcome acquired lapatinib resistance, and identify potential novel treatment strategies, with potential biomarkers, for HER2-positive breast cancer that is resistant to HER2 targeted therapies. Electronic supplementary material The online version of this article (10.1186/s12885-018-4852-1) contains supplementary material, which is available to authorized users.
- Published
- 2018
- Full Text
- View/download PDF
34. The HSP90 inhibitor NVP-AUY922 inhibits growth of HER2 positive and trastuzumab-resistant breast cancer cells
- Author
-
Alex J Eustace, Neil T Conlon, Denis M. Collins, Alexandra Canonici, Zulfiqar Qadir, John Crown, Neil A. O'Brien, Naomi Walsh, and Norma O'Donovan
- Subjects
0301 basic medicine ,Receptor, ErbB-2 ,medicine.medical_treatment ,Breast Neoplasms ,Hsp90 inhibitor ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,immune system diseases ,Trastuzumab ,Cell Line, Tumor ,medicine ,Humans ,Pharmacology (medical) ,HSP90 Heat-Shock Proteins ,skin and connective tissue diseases ,neoplasms ,Pharmacology ,Chemotherapy ,biology ,business.industry ,virus diseases ,Isoxazoles ,Resorcinols ,medicine.disease ,Hsp90 ,In vitro ,3. Good health ,030104 developmental biology ,Oncology ,Cell culture ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Female ,Breast cancer cells ,business ,medicine.drug - Abstract
As HER2 is a client protein of the molecular chaperone Hsp90, targeting Hsp90 may be beneficial in HER2-positive breast cancer. In this study, the activity of the Hsp90 inhibitor NVP-AUY922 was assessed in HER2 overexpressing breast cancer cell lines, including two cell line models of acquired trastuzumab-resistance. The seven HER2-positive breast cancer cell lines tested showed significant sensitivity to NVP-AUY922 in vitro, with IC50 values between 6 and 17 nM. Combining NVP-AUY922 with chemotherapy did not improve response. NVP-AUY922 in combination with trastuzumab, significantly enhanced growth inhibition in three of the seven cell lines tested. In conclusion, our data shows that NVP-AUY922 displays potent anti-cancer activity in HER2-positive and trastuzumab-resistant breast cancer cells, and supports further testing of NVP-AUY922 in patients with HER2-positive breast cancer.
- Published
- 2018
- Full Text
- View/download PDF
35. Mutant p53 as a therapeutic target for the treatment of triple-negative breast cancer: Preclinical investigation with the anti-p53 drug, PK11007
- Author
-
William M. Gallagher, Stephen F. Madden, Darran P. O'Connor, Norma O'Donovan, Matthias R. Bauer, Alan R. Fersht, John Crown, Naoise C Synnott, Michael J. Duffy, Alyson Murray, and Rut Klinger
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,Estrogen receptor ,Antineoplastic Agents ,Apoptosis ,Triple Negative Breast Neoplasms ,Biology ,Targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Cell Line, Tumor ,medicine ,Humans ,Sulfones ,Triple-negative breast cancer ,Cell Proliferation ,Cell growth ,Gene Expression Profiling ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Gene Ontology ,Pyrimidines ,030104 developmental biology ,Oncology ,Cell culture ,030220 oncology & carcinogenesis ,Immunology ,MCF-7 Cells ,Cancer research ,Female ,Mutant Proteins ,Tumor Suppressor Protein p53 ,Signal transduction - Abstract
The identification of a targeted therapy for patients with triple-negative breast cancer (TNBC) is one of the most urgent needs in breast cancer therapeutics. The p53 gene is mutated in approximately 80% of patients with TNBC, and is a potential therapeutic target for patients with this form of breast cancer. The 2-sulfonylpyrimidine compound, PK11007, preferentially decreases viability in p53-compromised cancer cell lines. We investigated PK11007 as a potential new treatment for TNBC. IC50 values for inhibition of proliferation in a panel of 17 breast cell lines by PK11007 ranged from 2.3 to 42.2 μM. There were significantly lower IC50 values for TNBC than for non-TNBC cell lines (p = 0.03) and for p53-mutated cell lines compared with p53 WT cells (p = 0.003). Response to PK11007 however, was independent of the estrogen receptor (ER) or HER2 status of the cell lines. In addition to inhibiting cell proliferation, PK11007 induced apoptosis in p53 mutant cell lines. Using RNAseq and gene ontology analysis, we found that PK11007 altered the expression of genes enriched in pathways involved in regulated cell death, regulation of apoptosis, signal transduction, protein refolding and locomotion. The observations that PK11007 inhibited cell proliferation, induced apoptosis and altered genes involved in cell death are all consistent with the ability of PK11007 to reactivate mutant p53. Based on our data, we conclude that targeting mutant p53 with PK11007 is a potential approach for treating p53-mutated breast cancer, including the subgroup with TN disease.
- Published
- 2018
- Full Text
- View/download PDF
36. The synthesis, structural characterization and biological evaluation of novel N-{para-(ferrocenyl) ethynyl benzoyl} amino acid and dipeptide methyl and ethyl esters as anticancer agents
- Author
-
Andy G. Harry, James P. Murphy, Dilip K. Rai, John Crown, Peter T.M. Kenny, and Norma O'Donovan
- Subjects
chemistry.chemical_classification ,Dipeptide ,integumentary system ,010405 organic chemistry ,Hydrochloride ,Stereochemistry ,Electrospray ionization ,Bioorganometallic chemistry ,Organic Chemistry ,010402 general chemistry ,01 natural sciences ,Biochemistry ,0104 chemical sciences ,Amino acid ,Inorganic Chemistry ,chemistry.chemical_compound ,chemistry ,Ferrocene ,otorhinolaryngologic diseases ,Materials Chemistry ,Proton NMR ,Physical and Theoretical Chemistry ,Benzoic acid - Abstract
A series of N -{ para -(ferrocenyl) ethynyl benzoyl} amino acid and dipeptide methyl and ethyl esters 4–18 were prepared by coupling para -(ferrocenyl) ethynyl benzoic acid 3 to the amino acids GABA(OMe), GABA(OEt) and the dipeptide esters GlyGly(OMe), GlyGly(OEt), Gly-L-Ala(OMe), Gly-L-Ala(OEt), Gly-D-Ala(OMe), Gly-D-Ala(OEt), Gly-L-Leu(OEt), Gly-L-Phe(OEt), SarGly(OMe), SarGly(OEt), Sar-L-Ala(OEt), L-ProGly(OEt) and L-Pro-L-Ala(OEt) using the standard N -(3-dimethylaminopropyl)- N’ -ethylcarbodiimide hydrochloride (EDC), 1-hydroxybenzotriazole (HOBt) protocol. All the compounds were fully characterized using a combination of I.R., UV-Vis, 1 H NMR, 13 C NMR, DEPT-135, 1 H- 13 C COSY (HMQC) spectroscopy and electrospray ionization mass spectrometry (ESI-MS). Selected compounds 5 , 7 , 9, 11, 16, 17 and 18 showed micromolar activity in the H1299 NSCLC cell line, with IC 50 values in the range of 3.8–8.3 μM.
- Published
- 2017
- Full Text
- View/download PDF
37. Dual inhibition of IGF1R and ER enhances response to trastuzumab in HER2 positive breast cancer cells
- Author
-
Stephen F. Madden, Neil A. O'Brien, Brigid C. Browne, Norma O'Donovan, John Crown, Laura Ivers, Martina McDermott, and Alexandra Canonici
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Receptor, ErbB-2 ,medicine.medical_treatment ,IGF1R Positive ,Breast Neoplasms ,Disease-Free Survival ,Receptor, IGF Type 1 ,Targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Trastuzumab ,Cell Line, Tumor ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Medicine ,Pyrroles ,skin and connective tissue diseases ,neoplasms ,Cell Proliferation ,Chemotherapy ,Oncogene ,business.industry ,Estrogen Receptor alpha ,Receptors, Somatomedin ,Cell cycle ,medicine.disease ,Xenograft Model Antitumor Assays ,Gene Expression Regulation, Neoplastic ,body regions ,Tamoxifen ,Pyrimidines ,030104 developmental biology ,030220 oncology & carcinogenesis ,Female ,business ,medicine.drug - Abstract
Although HER2 targeted therapies have improved prognosis for HER2 positive breast cancer, HER2 positive cancers which co-express ER have poorer response rates to standard HER2 targeted therapies, combined with chemotherapy, than HER2 positive/ER negative breast cancer. Administration of hormone therapy concurrently with chemotherapy and HER2 targeted therapy is generally not recommended. Using publically available gene expression datasets we found that high expression of IGF1R is associated with shorter disease-free survival in patients whose tumors are ER positive and HER2 positive. IGF1R is frequently expressed in HER2 positive breast cancer and there is significant evidence for crosstalk between IGF1R and both HER2 and ER. Therefore, we evaluated the therapeutic potential of targeting ER and IGF1R in cell line models of HER2/ER/IGF1R positive breast cancer, using tamoxifen and two IGF1R targeted tyrosine kinase inhibitors (NVP-AEW541 and BMS-536924). Dual inhibition of ER and IGF1R enhanced growth inhibition in the four HER2 positive cell lines tested and caused an increase in cell cycle arrest in G1 in BT474 cells. In addition, combined treatment with trastuzumab, tamoxifen and either of the IGF1R TKIs enhanced response compared to dual targeting strategies in three of the four HER2 positive breast cancer cell lines tested. Furthermore, in a cell line model of trastuzumab-resistant HER2 positive breast cancer (BT474/Tr), tamoxifen combined with an IGF1R TKI produced a similar enhanced response as observed in the parental BT474 cells suggesting that this combination may overcome acquired trastuzumab resistance in this model. Combining ER and IGF1R targeting with HER2 targeted therapies may be an alternative to HER2 targeted therapy and chemotherapy for patients with HER2/ER/IGF1R positive breast cancer.
- Published
- 2017
- Full Text
- View/download PDF
38. Vitamin D analogues: Potential use in cancer treatment
- Author
-
John Crown, Michael J. Duffy, Norma O'Donovan, Naoise C Synnott, and Alyson Murray
- Subjects
0301 basic medicine ,Paricalcitol ,Calcitriol ,business.industry ,Cancer ,Hematology ,Pharmacology ,medicine.disease ,Calcitriol receptor ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Breast cancer ,Oncology ,Neoplasms ,030220 oncology & carcinogenesis ,Toxicity ,medicine ,Vitamin D and neurology ,Animals ,Humans ,Vitamin D ,business ,Receptor ,medicine.drug - Abstract
The vitamin D receptor (VDR) is a member of the thyroid-steroid family of nuclear transcription factors. Following binding of the active form of vitamin D, i.e., 1,25(OH)2D3 (also known as calcitriol) and interaction with co-activators and co-repressors, VDR regulates the expression of several different genes. Although relatively little work has been carried out on VDR in human cancers, several epidemiological studies suggest that low circulating levels of vitamin D are associated with both an increased risk of developing specific cancer types and poor outcome in patients with specific diagnosed cancers. These associations apply especially in colorectal and breast cancer. Consistent with these findings, calcitriol as well as several of its synthetic analogues have been shown to inhibit tumor cell growth in vitro and in diverse animal model systems. Indeed, some of these vitamin D analogues with low calcemic inducing activity (e.g., EB1089, inecalcitol, paricalcitol) have progressed to clinical trials in patients with cancer. Preliminary results from these trials suggest that these vitamin D analogues have minimal toxicity, but clear evidence of efficacy remains to be shown. Although evidence of efficacy for mono-treatment with vitamin D analogues is currently lacking, several studies have reported that supplementation with calcitriol or the presence of high endogenous circulating levels of vitamin D enhances response to standard therapies.
- Published
- 2017
- Full Text
- View/download PDF
39. Pilot study of bevacizumab in combination with docetaxel and cyclophosphamide as adjuvant treatment for patients with early stage HER-2 negative breast cancer, including analysis of candidate circulating markers of cardiac toxicity: ICORG 08–10 trial
- Author
-
John McCaffrey, Mairin Rafferty, Michael J. Kennedy, M. Keane, Miriam O'Connor, Ashwini Maratha, Enda W. McDermott, Paul Donnellan, Oscar Breathhnach, D. Tryfonopoulos, John Crown, Paula Calvert, Rajnish Gupta, Giuseppe Gullo, Verena Murphy, Denis M. Collins, G. Leonard, Annette T. Byrne, Imelda Parker, Kathleen Scott, David W. Murray, Bonnie Ky, Michael J. Martin, Patrick Dicker, Alexandra Canonici, Norma O'Donovan, Liam Grogan, Andres Hernando, Alex J Eustace, Brian Moulton, Alice C. O’Farrell, William M. Gallagher, and Janice M. Walshe
- Subjects
Oncology ,medicine.medical_specialty ,Bevacizumab ,Cyclophosphamide ,medicine.medical_treatment ,bevacizumab ,docetaxel/cyclophosphamide ,lcsh:RC254-282 ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,breast cancer ,Internal medicine ,medicine ,Stage (cooking) ,030304 developmental biology ,Original Research ,0303 health sciences ,Chemotherapy ,business.industry ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Metastatic breast cancer ,3. Good health ,Docetaxel ,030220 oncology & carcinogenesis ,cardiotoxicity biomarker ,business ,Adjuvant ,medicine.drug - Abstract
Background: Combining bevacizumab and chemotherapy produced superior response rates compared with chemotherapy alone in metastatic breast cancer. As bevacizumab may cause hypertension (HTN) and increase the risk of cardiac failure, we performed a pilot study to evaluate the feasibility and toxicity of a non-anthracycline-containing combination of docetaxel with cyclophosphamide and bevacizumab in early stage breast cancer patients. Methods: Treatment consisted of four 3-weekly cycles of docetaxel and cyclophosphamide (75/600 mg/m2). Bevacizumab was administered 15 mg/kg intravenously on day 1, and then every 3 weeks to a total of 18 cycles of treatment. Serum biomarker concentrations of vascular endothelial growth factor (VEGF), cardiac troponin-I (cTnI), myeloperoxidase (MPO), and placental growth factor (PlGF) were quantified using enzyme-linked immunosorbent assay (ELISA) in 62 patients at baseline and whilst on treatment to determine their utility as biomarkers of cardiotoxicity, indicated by left ventricular ejection fraction (LVEF). Results: A total of 106 patients were accrued in nine sites. Median follow up was 65 months (1–72 months). Seventeen protocol-defined relapse events were observed, accounting for an overall disease-free survival (DFS) rate of 84%. The DFS rates for hormone receptor positive (HR+) and triple-negative (TN) patients were 95% versus 43%, respectively. The median time to relapse was 25 (12–54) months in TN patients versus 38 (22–71) months in HR+ patients. There have been 13 deaths related to breast cancer . The overall survival (OS) rate was 88%. The 5-year OS rate in HR+ versus TN was 95% versus 57%. None of the measured biomarkers predicted the development of cardiotoxicity. Conclusions: We observed a low relapse rate in node-positive, HR+ patients; however, results in TN breast cancer were less encouraging. Given the negative results of three large phase III trials, it is unlikely that this approach will be investigated further. Trial Registration ClinicalTrials.gov Identifier: NCT00911716.
- Published
- 2019
40. Dasatinib Treatment Increases Sensitivity to c-Met Inhibition in Triple-Negative Breast Cancer Cells
- Author
-
Patricia Gaule, B. Corkery, Kathy Gately, Michael J. Duffy, John Crown, Naomi Walsh, Nupur Mukherjee, Robert O'Connor, Norma O'Donovan, Sandra Roche, Alex J Eustace, and Kenneth J. O'Byrne
- Subjects
0301 basic medicine ,Cancer Research ,C-Met ,cMet ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,hemic and lymphatic diseases ,medicine ,IC50 ,Triple-negative breast cancer ,Kinase ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,In vitro ,3. Good health ,Dasatinib ,030104 developmental biology ,Oncology ,chemistry ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,Src kinase ,basal-like breast cancer ,Proto-oncogene tyrosine-protein kinase Src ,medicine.drug - Abstract
In pre-clinical studies, triple-negative breast cancer (TNBC) cells have demonstrated sensitivity to the multi-targeted kinase inhibitor dasatinib, however, clinical trials with single-agent dasatinib showed limited efficacy in unselected populations of breast cancer, including TNBC. To study potential mechanisms of resistance to dasatinib in TNBC, we established a cell line model of acquired dasatinib resistance (231-DasB). Following an approximately three-month exposure to incrementally increasing concentrations of dasatinib (200 nM to 500 nM) dasatinib, 231-DasB cells were resistant to the agent with a dasatinib IC50 value greater than 5 &mu, M compared to 0.04 ±, 0.001 µ, M in the parental MDA-MB-231 cells. 231-DasB cells also showed resistance (2.2-fold) to the Src kinase inhibitor PD180970. Treatment of 231-DasB cells with dasatinib did not inhibit phosphorylation of Src kinase. The 231-DasB cells also had significantly increased levels of p-Met compared to the parental MDA-MB-231 cells, as measured by luminex, and resistant cells demonstrated a significant increase in sensitivity to the c-Met inhibitor, CpdA, with an IC50 value of 1.4 ±, 0.5 µ, M compared to an IC50 of 6.8 ±, 0.2 µ, M in the parental MDA-MB-231 cells. Treatment with CpdA decreased p-Met and p-Src in both 231-DasB and MDA-MB-231 cells. Combined treatment with dasatinib and CpdA significantly inhibited the growth of MDA-MB-231 parental cells and prevented the emergence of dasatinib resistance. If these in vitro findings can be extrapolated to human cancer treatment, combined treatment with dasatinib and a c-Met inhibitor may block the development of acquired resistance and improve response rates to dasatinib treatment in TNBC.
- Published
- 2019
41. Combined targeting EGFR and SRC as a potential novel therapeutic approach for the treatment of triple negative breast cancer
- Author
-
Kevin P. Fanning, A. Browne, Alexandra Canonici, Mattia Cremona, Mohamed F. K. Ibrahim, Alex J Eustace, Flavio Solca, John Crown, Fiona O'Neill, Justine Meiller, Norma O'Donovan, Clare Morgan, Neil T Conlon, Sandra Roche, and Bryan T. Hennessy
- Subjects
Bcl2 ,Afatinib ,TNBC in 2019: Promising Signals for the Treatment of a Formidable Disease ,EGFR ,afatinib ,lcsh:RC254-282 ,Therapeutic approach ,Breast cancer ,hemic and lymphatic diseases ,medicine ,dasatinib ,Epidermal growth factor receptor ,Triple-negative breast cancer ,Original Research ,biology ,business.industry ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Dasatinib ,Oncology ,Cancer research ,biology.protein ,business ,TNBC ,medicine.drug ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Background:Triple negative breast cancer (TNBC) is an aggressive subtype of breast cancer with limited therapeutic options. Epidermal growth factor receptor (EGFR) has been shown to be over-expressed in TNBC and represents a rational treatment target.Methods:We examined single agent and combination effects for afatinib and dasatinib in TNBC. We then determined IC50and combination index values using Calcusyn. Functional analysis of single and combination treatments was performed using reverse phase protein array and cell cycle analysis. Finally, we determined the anticancer effects of the combination in vivo.Results:A total of 14 TNBC cell lines responded to afatinib with IC50values ranging from 0.008 to 5.0 µM. Three cell lines, belonging to the basal-like subtype of TNBC, were sensitive to afatinib. The addition of afatinib enhanced response to the five other targeted therapies in HCC1937 and HDQP1 cells. The combination of afatinib with dasatinib caused the greatest growth inhibition in both cell lines. The afatinib/dasatinib combination was synergistic and/or additive in 13/14 TNBC cell lines. Combined afatinib/dasatinib treatment induced G1 cell cycle arrest. Reverse phase protein array results showed the afatinib/dasatinib combination resulted in efficient inhibition of both pERK(T202/T204) and pAkt(S473) signalling in BT20 cells, which was associated with the greatest antiproliferative effects. High baseline levels of pSrc(Y416) and pMAPK(p38) correlated with sensitivity to afatinib, whereas low levels of B-cell lymphoma 2 (Bcl2) and mammalian target of rapamycin (mTOR) correlated with synergistic growth inhibition by combined afatinib and dasatinib treatment. In vivo, the combination treatment inhibited tumour growth in a HCC1806 xenograft model.Conclusions:We demonstrate that afatinib combined with dasatinib has potential clinical activity in TNBC but warrants further preclinical investigation.
- Published
- 2019
42. HER2-Targeted Tyrosine Kinase Inhibitors Cause Therapy-Induced-Senescence in Breast Cancer Cells
- Author
-
Neil T Conlon, Brigid C. Browne, Naoise C Synnott, Michael J. Duffy, Adam Szabo, Norma O'Donovan, M McDermott, Neil A. O'Brien, and John Crown
- Subjects
0301 basic medicine ,Senescence ,Cancer Research ,Programmed cell death ,senescence ,Afatinib ,Oncology and Carcinogenesis ,afatinib ,Biology ,Lapatinib ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,0302 clinical medicine ,breast cancer ,HER2 ,medicine ,2.1 Biological and endogenous factors ,Aetiology ,lapatinib ,skin and connective tissue diseases ,Cancer ,Transfection ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,TKI ,erbB2 ,3. Good health ,respiratory tract diseases ,neratinib ,trastuzumab ,030104 developmental biology ,Oncology ,Cell culture ,5.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Neratinib ,Cancer research ,Development of treatments and therapeutic interventions ,Tyrosine kinase ,medicine.drug - Abstract
Prolonged treatment of HER2 positive breast cancer cells with tyrosine kinase inhibitors (TKIs) leads to the emergence of acquired resistance. However, the effects of continuous TKI exposure on cell fate, and the steps leading to the acquisition of a resistant phenotype are poorly understood. To explore this, we exposed five HER2 positive cells lines to HER2 targeted therapies for periods of up to 4 weeks and examined senescence associated &beta, galactosidase (SA-&beta, gal) activity together with additional markers of senescence. We found that lapatinib treatment resulted in phenotypic alterations consistent with a senescent phenotype and strong SA-&beta, gal activity in HER2-positive cell lines. Lapatinib-induced senescence was associated with elevated levels of p15 and p27 but was not dependent on the expression of p16 or p21. Restoring wild type p53 activity either by transfection or by treatment with APR-246, a molecule which reactivates mutant p53, blocked lapatinib-induced senescence and caused increased cell death. In contrast to lapatinib, SA-&beta, gal activity was not induced by exposing the cells to trastuzumab as a single agent but co-administration of lapatinib and trastuzumab induced senescence, as did treatment of the cells with the irreversible HER2 TKIs neratinib and afatinib. Neratinib- and afatinib-induced senescence was not reversed by removing the drug whereas lapatinib-induced senescence was reversible. In summary, therapy-induced senescence represents a novel mechanism of action of HER2 targeting agents and may be a potential pathway for the emergence of resistance.
- Published
- 2019
- Full Text
- View/download PDF
43. Validated biomarkers: The key to precision treatment in patients with breast cancer
- Author
-
Michael J. Duffy, Enda W. McDermott, Norma O'Donovan, and J.P. Crown
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Receptor, ErbB-2 ,medicine.medical_treatment ,Estrogen receptor ,Breast Neoplasms ,Systemic therapy ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,MammaPrint ,Internal medicine ,Plasminogen Activator Inhibitor 1 ,Biomarkers, Tumor ,medicine ,Humans ,Precision Medicine ,Gynecology ,Chemotherapy ,medicine.diagnostic_test ,business.industry ,General Medicine ,Prognosis ,medicine.disease ,Precision medicine ,030104 developmental biology ,Receptors, Estrogen ,030220 oncology & carcinogenesis ,Biomarker (medicine) ,Female ,Surgery ,Receptors, Progesterone ,business ,Oncotype DX - Abstract
Recent DNA sequencing and gene expression studies have shown that at a molecular level, almost every case of breast cancer is unique and different from other breast cancers. For optimum management therefore, every patient should receive treatment that is guided by the molecular composition of their tumor, i.e., precision treatment. While such a scenario is still some distance into the future, biomarkers are beginning to play an important role in preparing the way for precision treatment. In particular, biomarkers are increasingly being used for predicting patient outcome and informing as to the most appropriate type of systemic therapy to be administered. Mandatory biomarkers for every newly diagnosed case of breast cancer are estrogen receptors and progesterone receptors in selecting patients for endocrine treatment and HER2 for identifying patients likely to benefit from anti-HER2 therapy. Amongst the best validated prognostic biomarker tests are uPA/PAI-1, MammaPrint and Oncotype DX. Although currently, there are no biomarkers available for predicting response to specific forms of chemotherapy, uPA/PAI-1 and Oncotype DX can aid the identification of lymph node-negative patients that are most likely to benefit from adjuvant chemotherapy, in general. In order to accelerate progress towards precision treatment for women with breast cancer, we need additional predictive biomarkers, especially for enhancing the positive predictive value for endocrine and anti-HER2 therapies, as well as biomarkers for predicting response to specific forms of chemotherapy. The ultimate biomarker test for achieving the goal of precision treatment for patients with breast cancer will likely require a combination of gene sequencing and transcriptomic analysis of every patient's tumor.
- Published
- 2016
- Full Text
- View/download PDF
44. Mutant p53: a novel target for the treatment of patients with triple-negative breast cancer?
- Author
-
Michael J. Duffy, William M. Gallagher, Alyson Murray, Maeve Kiely, Naoise C Synnott, Norma O'Donovan, Patrick A. Kiely, Darran P. O'Connor, Patricia M. McGowan, and John Crown
- Subjects
0301 basic medicine ,Cancer Research ,Cell growth ,business.industry ,medicine.medical_treatment ,Mutant ,CA 15-3 ,medicine.disease ,humanities ,Targeted therapy ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Breast cancer ,Oncology ,Apoptosis ,Cell culture ,030220 oncology & carcinogenesis ,Immunology ,medicine ,Cancer research ,business ,Triple-negative breast cancer - Abstract
The identification and validation of a targeted therapy for patients with triple-negative breast cancer (TNBC) is currently one of the most urgent needs in breast cancer therapeutics. One of the key reasons for the failure to develop a new therapy for this subgroup of breast cancer patients has been the difficulty in identifying a highly prevalent, targetable molecular alteration in these tumors. Recently however, the p53 gene was found to be mutated in approximately 80% of basal/TNBC, raising the possibility that targeting the mutant p53 protein product might be a new approach for the treatment of this form of breast cancer. In this study, we investigated the anti-cancer activity of PRIMA-1 and PRIMA-1MET (APR-246), two compounds which were previously reported to reactivate mutant p53 and convert it to a form with wild-type (WT) properties. Using a panel of 18 breast cancer cell lines and 2 immortalized breast cell lines, inhibition of proliferation by PRIMA-1 and PRIMA-1MET was found to be cell-line dependent, but independent of cell line molecular subtype. Although response was independent of molecular subtype, p53 mutated cell lines were significantly more sensitive to PRIMA-1MET than p53 WT cells (p = 0.029). Furthermore, response (measured as IC50 value) correlated significantly with p53 protein level as measured by ELISA (p = 0.0089, r=-0.57, n = 19). In addition to inhibiting cell proliferation, PRIMA-1MET induced apoptosis and inhibited migration in a p53 mutant-dependent manner. Based on our data, we conclude that targeting mutant p53 with PRIMA-1MET is a potential new approach for treating p53-mutated breast cancer, including the subgroup with triple-negative (TN) disease.
- Published
- 2016
- Full Text
- View/download PDF
45. Abstract CT212: A phase III randomized study of paclitaxel (T) and trastuzumab (H) versus T, H and lapatinib (L) in first line treatment of HER2+ metastatic breast cancer (MBC)
- Author
-
Johanna Mattson, Pedro Sánchez Rovira, Álvaro Rodríguez Lescure, Norma O'Donovan, M. Keane, Catherine Kelly, John Crown, Seamus O'Reilly, Raquel Andrés Conejero, Linda Coate, Andres Hernando, Miriam O'Connor, Elena Alvarez, Imelda Parker, Anna Barbro Sætersdal, Luis Costa, Manuel Ramos Vazquez, Denis M. Collins, Laura López, Coralia Bueno Muino, Blanca Cantos Sanchez De Ibarguen, Brian Moulton, Miguel Martín, Bryan T. Hennessy, William Jacot, Esperanza J. Carcache de Blanco, Margarida Brito, Marc Nolan, M. John Kennedy, and Giuseppe Gullo
- Subjects
0303 health sciences ,Cancer Research ,medicine.medical_specialty ,Chemotherapy ,business.industry ,medicine.medical_treatment ,Cancer ,Lapatinib ,medicine.disease ,Metastatic breast cancer ,Gastroenterology ,3. Good health ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Oncology ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Hormonal therapy ,Progression-free survival ,business ,030304 developmental biology ,medicine.drug - Abstract
Background: The addition of H to chemotherapy improved disease outcome in HER2+ MBC. The combination of H and L with chemotherapy improved event free survival (EFS) in the neo-adjuvant setting but not in the adjuvant setting. However, improved EFS did not translate to overall survival benefit. This international trial (ICORG (CTRIAL-IE) 11-10/NCT01526369) compared the efficacy of TH vs. THL in first line treatment of HER2+ MBC. The primary aim of this study was to assess any benefit for the addition of L to TH in an advanced HER2+ breast cancer patient population. Target accrual was not met due to changes to standard of care mid-study. Methods: Patients (Pts) (n= 75 enrolled, randomized 1:1) received weekly T (80mg/m², for 3 weeks of a 4 week cycle) + H (8mg/kg loading dose on cycle 1 day 1 and 4mg/kg every 2 weeks) + L (1,000 mg daily)) until disease progression, unacceptable toxicity or consent withdrawal. The primary outcome measure was progression free survival (PFS). Secondary outcome measure was overall survival (OS). Eligibility criteria included: Female > 18 years, invasive MBC, measurable disease by RECIST criteria V1.1, HER2+ by testing of the primary tumour and if available the biopsied metastatic lesion. No prior systemic therapy for metastatic disease (except one line of hormonal therapy for metastatic disease without H). No recurrence within 12 months (mo) from completion of adjuvant chemotherapy to the development of metastatic disease. No recurrence within 6 mo from completion of adjuvant H to the development of metastatic disease and no prior L treatment. Results: Of the 75 pts enrolled, n=65 (TH n=34, THL n=31) were included in the PFS analysis. Median PFS (95% CI) was numerically higher, but not statistically significant, in THL (24.0 mo (9.5, 34.5)) vs TH (19.3 mo (8.7, 22.7)), HR 0.827, p= 0.601. For OS analysis, n=69 (TH n=35 and THL n=34) were included. 41/69 (59.4%) had no prior therapy. No statistically significant, clinically meaningful improvement in OS (Median (95% CI)) was observed (TH 53.8 mo (41.8, -) vs. THL 45.6 mo (29.7, -), HR 1.048, p=0.891). Analysis of the safety set (n=70 (TH n= 36, THL n=34)) revealed a higher incidence of Grade 3/4 AEs in THL (61.8%) vs. TH (50%), p=0.322. There were 2 on-treatment fatalities due to adverse events (AEs) in the THL arm, one due to respiratory failure, the other due to central nervous system metastasis. There were no on-treatment fatalities in the TH arm. GI disorders were the most prevalent AE, with the incidence of diarrhea significantly higher in the THL arm (88% vs. 42%, p Citation Format: John Crown, William Jacot, Denis M. Collins, Linda Coate, Anna Saetersdal, M John Kennedy, Catherine Kelly, Blanca Cantos Sanchez De Ibarguen, Raquel Andrés Conejero, Luis Costa, Margarida Brito, Maccon Keane, Pedro Sanchez Rovira, Miguel Martin, Miriam O'Connor, Manuel Ramos Vazquez, Elena Alvarez, Seamus O'Reilly, Johanna Mattson, Laura Jolis Lopez, Alvaro Rodriguez Lescure, Esperanza Blanco, Coralia Bueno Muino, Brian Moulton, Norma O'Donovan, Andres Hernando, Marc Nolan, Imelda Parker, Giuseppe Gullo, Bryan Hennessy. A phase III randomized study of paclitaxel (T) and trastuzumab (H) versus T, H and lapatinib (L) in first line treatment of HER2+ metastatic breast cancer (MBC) [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr CT212.
- Published
- 2020
- Full Text
- View/download PDF
46. Abstract 951: The effect of neo-adjuvant chemotherapy on immune cell phenotype and cytotoxic capacity in HER2+ breast cancer patients
- Author
-
Jean M. Fletcher, Alfonso Blanco, Barry Moran, N. Gaynor, John Crown, Martina McDermott, Denis M. Collins, Norma O'Donovan, Alex J Eustace, and Alexandra Canonici
- Subjects
Cancer Research ,Cell phenotype ,Immune system ,Breast cancer ,Oncology ,business.industry ,Cancer research ,Cytotoxic T cell ,Medicine ,business ,Neo adjuvant chemotherapy ,medicine.disease - Abstract
Introduction: The phase II neoadjuvant clinical trial ICORG10-05 (NCT01485926) compared chemotherapy (docetaxel, carboplatin) in combination with trastuzumab, trastuzumab and lapatinib or lapatinib alone in HER2+ breast cancer patients. Lapatinib is a dual HER2/EGFR tyrosine kinase inhibitor. Trastuzumab is a monoclonal antibody which targets HER2 and is capable of eliciting antibody-dependent cell-mediated cytotoxicity (ADCC) mediated by immune effector cells. Studies have shown that tumour infiltrating lymphocyte (TIL) levels and the cytotoxic capacity of circulating immune cells can correlate with response to trastuzumab. Less is known regarding the effect of chemotherapy on the immune response to trastuzumab. This study examines the effects of neo-adjuvant chemotherapy on the phenotype and cytotoxic capacity of peripheral blood mononuclear cells (PBMCs) of HER2+ breast cancer patients. Methods: Matched blood samples were taken pre- and post-neoadjuvant treatment. PBMCs were isolated by density centrifugation and frozen. Direct PBMC-mediated cytotoxicity and trastuzumab-ADCC levels were assessed against the HER2+ breast cancer cell line (SKBR3) and non-MHC class I-restricted leukemic cell line (K562) using a FACS based assay (n=19 matched pre-and post-treatment samples). The immunophenotype of 17 pre-treatment and 13 post-treatment samples was also determined using the DURAClone IM antibody panel (CD16, CD56, CD19, CD14, CD4, CD8, CD3, CD45) on the CytoFLEX platform. Analysis of both data sets was performed using FCS Express and MedCalc. Results: The cytotoxic capacity of PBMCs was reduced following neo-adjuvant chemotherapy. Direct cytotoxicity elicited against the K562 cell line was reduced post-treatment (p=0.04). ADCC elicited against the SKBR3 cell line was also decreased in post-treatment samples (p=0.009). A decrease in cytotoxic capacity was associated with alterations in the immunophenotype of the post-treatment PBMC samples. The proportion of CD56+ NK cells (p=0.001), CD19+ B cells (p=0.001) and CD14+ monocytes (p=0.03) decreased significantly post-treatment. CD56+ NK cells are hypothesised to be the main ADCC effector cell population in peripheral blood. Interestingly, the proportion of CD3+ T cells (p=0.002), including CD4+ (p=0.007) and CD8+ (p=0.035) T cell populations, were increased post-treatment. Conclusion: These results indicate that neo-adjuvant chemotherapy reduces the cytotoxic capacity of circulating immune cells and alters the proportion of adaptive and innate immune cell subsets. These effects warrant further investigation particularly with the emergence of immunotherapies in HER2+ breast cancer. Citation Format: Nicola Gaynor, Alfonso Blanco, Alexandra Canonici, Alexander J. Eustace, Martina McDermott, Barry Moran, Jean M. Fletcher, Norma O'Donovan, John Crown, Denis M. Collins. The effect of neo-adjuvant chemotherapy on immune cell phenotype and cytotoxic capacity in HER2+ breast cancer patients [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 951.
- Published
- 2020
- Full Text
- View/download PDF
47. Anti-PD-1 responsive circulating immune cells as biomarkers of response to neoadjuvant chemotherapy in HER2+ breast cancer (BC)
- Author
-
Denis M. Collins, Norma O'Donovan, N. Gaynor, and John Crown
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Chemotherapy ,business.industry ,medicine.medical_treatment ,Anti pd 1 ,chemical and pharmacologic phenomena ,medicine.disease ,Alternative treatment ,Breast cancer ,Immune system ,Internal medicine ,medicine ,skin and connective tissue diseases ,business ,Pathological ,Complete response - Abstract
e12646 Background: HER2+ BC patients (pts) achieving a pathological complete response (pCR) to neo-adjuvant therapy have better long-term survival outcomes. Alternative treatment strategies may be more beneficial for pts that will not achieve a pCR but there are no predictive biomarkers for stratification of pts to appropriate clinical studies. Antibody-dependent cell-mediated cytotoxicity (ADCC) mediated by immune cells like NK cells can be elicited by therapeutic antibody therapies including the HER2-targeting trastuzumab (T). We hypothesised that the presence of immune checkpoint-inhibited ADCC-mediating immune cells in the blood may identify pts with an immune-suppressed tumour microenvironment who will not achieve a pCR. We used an in vitro functional assay to assess the PD-1-inhibited ADCC response of pre-treatment peripheral blood mononuclear cells (PBMCs) from HER2+ BC pts as a potential biomarker of pCR. Methods: Pre-treatment PBMC samples (n = 21) were obtained from HER2+ BC pts who received neo-adjuvant chemotherapy (docetaxel, carboplatin) with a HER2-targeted therapy (T n = 5, T and lapatinib (L) n = 13, or L alone n = 3), ICORG 10-05 (NCT01485926). Pt response was determined as pCR (n = 7), partial response (PR, n = 10) or non-response (NR, n = 4). The immune function assay used a flow cytometry-based method to determine the T (10µg/µl)-mediated ADCC (T-ADCC) capacity of the PBMCs +/- the anti-PD-1 therapy pembrolizumab (P) (10µg/µl). The HER2+ BC cell line SKBR3 was used as the target cell line. Direct PBMC-mediated cytotoxicity against the non-MHC Class I-restricted leukemic K562 cell line was also determined. Statistical analysis was carried out using the MedCalc program. Results: There was no significant difference in direct cytotoxicity against K562 (pCR vs NR p = 0.13, pCR vs PR p = 0.52, PR vs NR p = 0.40), or in T-ADCC against SKBR3 (pCR vs NR p = 0.71, pCR vs PR p = 0.52, PR vs NR p = 0.32), between response cohorts in the absence of P. When T-ADCC was examined in individual pt samples, the addition of P resulted in significant increases in T-ADCC vs T-ADCC in the absence of P in 3/4 NR (p = 0.03, p = 0.01, p = 0.002) and 4/10 PR (p = 0.003, p = 0.007, p = 0.009, p = 0.01). No significant increase in T-ADCC was observed in the pCR cohort (0/7) as a result of the addition of P. Using pre-treatment PBMCs, the assay detected 7/14 (50%) of HER2+ BC pts who did not achieve a pCR. Conclusions: PD-1-inhibited ADCC-capable immune cells may identify a subset of HER2+ BC pts who will not achieve a pCR. Further investigation is warranted using a larger patient cohort.
- Published
- 2020
- Full Text
- View/download PDF
48. Systems modeling accurately predicts responses to genotoxic agents and their synergism with BCL-2 inhibitors in triple negative breast cancer cells
- Author
-
Andreas U. Lindner, Norma O'Donovan, Heiko Düssmann, Jochen H. M. Prehn, and Federico Lucantoni
- Subjects
0301 basic medicine ,Cancer Research ,Programmed cell death ,Systems Analysis ,Immunology ,Triple Negative Breast Neoplasms ,Article ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,medicine ,Humans ,MCL1 ,lcsh:QH573-671 ,Triple-negative breast cancer ,Cisplatin ,lcsh:Cytology ,Chemistry ,Venetoclax ,Antagonist ,Cell Biology ,3. Good health ,030104 developmental biology ,Paclitaxel ,Proto-Oncogene Proteins c-bcl-2 ,Apoptosis ,Cancer research ,medicine.drug - Abstract
Triple negative breast cancer (TNBC) is an aggressive form of breast cancer which accounts for 15–20% of this disease and is currently treated with genotoxic chemotherapy. The BCL2 (B-cell lymphoma 2) family of proteins controls the process of mitochondrial outer membrane permeabilization (MOMP), which is required for the activation of the mitochondrial apoptosis pathway in response to genotoxic agents. We previously developed a deterministic systems model of BCL2 protein interactions, DR_MOMP that calculates the sensitivity of cells to undergo mitochondrial apoptosis. Here we determined whether DR_MOMP predicts responses of TNBC cells to genotoxic agents and the re-sensitization of resistant cells by BCL2 inhibitors. Using absolute protein levels of BAX, BAK, BCL2, BCL(X)L and MCL1 as input for DR_MOMP, we found a strong correlation between model predictions and responses of a panel of TNBC cells to 24 and 48 h cisplatin (R2 = 0.96 and 0.95, respectively) and paclitaxel treatments (R2 = 0.94 and 0.95, respectively). This outperformed single protein correlations (best performer BCL(X)L with R2 of 0.69 and 0.50 for cisplatin and paclitaxel treatments, respectively) and BCL2 proteins ratio (R2 of 0.50 for cisplatin and 0.49 for paclitaxel). Next we performed synergy studies using the BCL2 selective antagonist Venetoclax /ABT199, the BCL(X)L selective antagonist WEHI-539, or the MCL1 selective antagonist A-1210477 in combination with cisplatin. In silico predictions by DR_MOMP revealed substantial differences in treatment responses of BCL(X)L, BCL2 or MCL1 inhibitors combinations with cisplatin that were successfully validated in cell lines. Our findings provide evidence that DR_MOMP predicts responses of TNBC cells to genotoxic therapy, and can aid in the choice of the optimal BCL2 protein antagonist for combination treatments of resistant cells.
- Published
- 2018
49. Preclinical evaluation targeting both IGF1R and IR in triple negative breast cancer
- Author
-
Justine Meiller, Sandra Roche, Fiona O'Neill, Norma O'Donovan, Stephen F. Madden, J.P. Crown, Alexandra Canonici, Denis M. Collins, Patricia Gaule, Alex J Eustace, Nupur Mukherjee, and Neil T Conlon
- Subjects
Oncology ,Linsitinib ,medicine.medical_specialty ,business.industry ,Hematology ,medicine.disease ,Tnbc cell ,Chemotherapy regimen ,chemistry.chemical_compound ,Breast cancer ,chemistry ,Docetaxel ,Growth factor receptor ,Internal medicine ,medicine ,business ,Triple-negative breast cancer ,medicine.drug ,Insulin-like growth factor 1 receptor - Abstract
Background Insulin Receptor (INSR) signalling may play a role in resistance to insulin-like growth factor receptor (IGF1R) therapy. Although IGF1R is frequently expressed in triple negative breast cancer (TNBC), we found that an IGF1R monoclonal antibody did not inhibit the growth of TNBC cells. Thus, we hypothesised that dual targeting of IGF1R and INSR may be more effective in TNBC. Methods Relative mRNA expression of INSR and IGFIR were analysed across different subtypes of breast cancer using the BreastMark database. INSR-A, INSR-B, IGF1R and insulin like growth factor 2 (IGFII) was quantified by qRT-PCR in a panel of 11 TNBC cell lines. Proliferation assays were performed on TNBC cell lines treated with the dual IR/INSR inhibitor Linsitinib (OSI-906). Combinations were performed by testing Linsitinib with Xentuzumab (BI-836845), cisplatin, docetaxel, or the PI3K inhibitor Dactolisib (NVP-BEZ235). We also tested the anti-proliferative effect of Linsitinib in combination with Xentuzumab when stimulated with IGF-I and IGF-II following serum-starvation for 24 hours. Finally, Linsitinib was tested in HCC-1143 xenografts to assess the effect of low-dose treatment on tumour formation. Results No significant differences were observed between the basal-like and non-basal-like cell lines for IR or IGF1R mRNA expression. However, IGF-II mRNA was more frequently detected in non-basal-like (80%, 4/5) compared to basal-like cell lines (20%, 1/5). Only three of the 12 cell lines tested showed sensitivity to Linsitinib with IC50 values less than 10 µM. The two most sensitive cell lines, HDQ-P1 and HCC1143, expressed detectable levels of IGF1R, INSR-A and INSR-B mRNAs. Although Linsitinib blocked IGF-I and IGF-II stimulated proliferation in both HCC1143 and HDQ-P1 cells, addition of Linsitinib to either chemotherapy or Dactolisib did not enhance therapeutic response. Linisitinib did not reduce tumour formation or tumour growth in an in vivo TNBC cell-line xenograft model. Conclusions Although IGF1R has been shown to be frequently expressed in TNBC, our in vitro and in vivo data suggest that it may not be a good therapeutic target in the TNBC subtype. Legal entity responsible for the study The authors. Funding Boehringer Ingleheim. Disclosure N. O’Donovan: Research grant / Funding (institution): Boehringer Ingleheim. J. Crown: Honoraria (self), Speaker Bureau / Expert testimony, Research grant / Funding (institution): Boehringer Ingleheim; Shareholder / Stockholder / Stock options, Full / Part-time employment: OncoMark Limited; Honoraria (self), Advisory / Consultancy, Speaker Bureau / Expert testimony, Research grant / Funding (institution): Eisai; Honoraria (self): Amgen; Honoraria (self), Advisory / Consultancy, Research grant / Funding (institution): Puma Biotechnology; Honoraria (self), Advisory / Consultancy: Seattle Genetics; Honoraria (self), Advisory / Consultancy, Speaker Bureau / Expert testimony, Travel / Accommodation / Expenses: Pfizer; Honoraria (self), Advisory / Consultancy: Vertex; Honoraria (self), Speaker Bureau / Expert testimony: Genomic Health; Honoraria (self), Advisory / Consultancy, Research grant / Funding (institution), Travel / Accommodation / Expenses: Roche; Honoraria (self), Travel / Accommodation / Expenses: MSD Oncology; Travel / Accommodation / Expenses: AstraZeneca; Travel / Accommodation / Expenses: Abbvie. All other authors have declared no conflicts of interest.
- Published
- 2019
- Full Text
- View/download PDF
50. Kinase inhibitor screening identifies CDK4 as a potential therapeutic target for melanoma
- Author
-
John Crown, Alex J Eustace, Thamir Mahgoub, Naomi Walsh, Norma O'Donovan, and Denis M. Collins
- Subjects
Neuroblastoma RAS viral oncogene homolog ,Cancer Research ,Indoles ,CDK4 ,Pyridines ,Antineoplastic Agents ,fascaplysin ,Piperazines ,Cyclin-dependent kinase ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,melanoma ,medicine ,Humans ,Molecular Targeted Therapy ,Neoplasm Metastasis ,Vemurafenib ,Clonogenic assay ,Protein Kinase Inhibitors ,neoplasms ,Cell Proliferation ,Sulfonamides ,cyclin dependent kinase ,biology ,Cell growth ,Cyclin-dependent kinase 4 ,Melanoma ,Cyclin-Dependent Kinase 4 ,Articles ,Cell cycle ,PD0332991 ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Oncology ,biology.protein ,Cancer research ,Drug Screening Assays, Antitumor ,kinase inhibitor library ,medicine.drug - Abstract
Despite recent advances in targeted therapies and immunotherapies metastatic melanoma remains only rarely curable. The objective of the present study was to identify novel therapeutic targets for metastatic melanoma. A library of 160 well-characterised and potent protein kinase inhibitors was screened in the BRAF mutant cell line Sk-Mel-28, and the NRAS mutant Sk-Mel-2, using proliferation assays. Of the 160 inhibitors tested, 20 achieved >50% growth inhibition in both cell lines. Six of the 20 were cyclin dependent kinase (CDK) inhibitors, including two CDK4 inhibitors. Fascaplysin, a synthetic CDK4 inhibitor, was further tested in 8 melanoma cell lines. The concentration of fascaplysin required to inhibit growth by 50% (IC50 value) ranged from 0.03 to 0.22 μM. Fascaplysin also inhibited clonogenic growth and induced apoptosis. Sensitivity to PD0332991, a therapeutic CDK4/6 inhibitor was also evaluated in the melanoma cell lines. PD0332991 IC50 values ranged from 0.13 to 2.29 μM. Similar to fascaplysin, PD0332991 inhibited clonogenic growth of melanoma cells and induced apoptosis. Higher levels of CDK4 protein correlated with lower sensitivity to PD0332991 in the cell lines. Combined treatment with PD0332991 and the BRAF inhibitor PLX4032, showed additive anti-proliferative effects in the BRAF mutant cell line Malme-3M. In summary, targeting CDK4 inhibits growth and induces apoptosis in melanoma cells in vitro, suggesting that CDK4 may be a rational therapeutic target for metastatic melanoma.
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.