Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Cornelia Quadt is active.

Publication


Featured researches published by Cornelia Quadt.


Nature | 2015

Convergent loss of PTEN leads to clinical resistance to a PI(3)Kα inhibitor

Dejan Juric; Pau Castel; Malachi Griffith; Obi L. Griffith; Helen H. Won; Haley Ellis; Saya H. Ebbesen; Benjamin J. Ainscough; Avinash Ramu; Gopa Iyer; Ronak Shah; Tiffany Huynh; Mari Mino-Kenudson; Dennis C. Sgroi; Steven J. Isakoff; Ashraf Thabet; Leila Elamine; David B. Solit; Scott W. Lowe; Cornelia Quadt; Malte Peters; Adnan Derti; Robert Schegel; Alan Huang; Elaine R. Mardis; Michael F. Berger; José Baselga; Maurizio Scaltriti

Broad and deep tumour genome sequencing has shed new light on tumour heterogeneity and provided important insights into the evolution of metastases arising from different clones. There is an additional layer of complexity, in that tumour evolution may be influenced by selective pressure provided by therapy, in a similar fashion to that occurring in infectious diseases. Here we studied tumour genomic evolution in a patient (index patient) with metastatic breast cancer bearing an activating PIK3CA (phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit alpha, PI(3)Kα) mutation. The patient was treated with the PI(3)Kα inhibitor BYL719, which achieved a lasting clinical response, but the patient eventually became resistant to this drug (emergence of lung metastases) and died shortly thereafter. A rapid autopsy was performed and material from a total of 14 metastatic sites was collected and sequenced. All metastatic lesions, when compared to the pre-treatment tumour, had a copy loss of PTEN (phosphatase and tensin homolog) and those lesions that became refractory to BYL719 had additional and different PTEN genetic alterations, resulting in the loss of PTEN expression. To put these results in context, we examined six other patients also treated with BYL719. Acquired bi-allelic loss of PTEN was found in one of these patients, whereas in two others PIK3CA mutations present in the primary tumour were no longer detected at the time of progression. To characterize our findings functionally, we examined the effects of PTEN knockdown in several preclinical models (both in cell lines intrinsically sensitive to BYL719 and in PTEN-null xenografts derived from our index patient), which we found resulted in resistance to BYL719, whereas simultaneous PI(3)K p110β blockade reverted this resistance phenotype. We conclude that parallel genetic evolution of separate metastatic sites with different PTEN genomic alterations leads to a convergent PTEN-null phenotype resistant to PI(3)Kα inhibition.


Science Translational Medicine | 2013

mTORC1 Inhibition Is Required for Sensitivity to PI3K p110α Inhibitors in PIK3CA-Mutant Breast Cancer

Moshe Elkabets; Sadhna Vora; Dejan Juric; Natasha Morse; Mari Mino-Kenudson; Taru A. Muranen; Jessica J. Tao; Ana Bosch Campos; Jordi Rodon; Yasir H. Ibrahim; Violeta Serra; Vanessa Rodrik-Outmezguine; Saswati Hazra; Sharat Singh; Phillip Kim; Cornelia Quadt; Manway Liu; Alan Huang; Neal Rosen; Jeffrey A. Engelman; Maurizio Scaltriti; José Baselga

Persistent mTORC1 signaling correlates with resistance to PI3K p110α inhibition in breast cancer, which can be overcome by inhibiting mTORC1. Caveat mTOR In recent years, numerous new drugs have been developed to take advantage of specific molecular changes in cancer cells. Unfortunately, tumors are often a step ahead of the scientists, becoming resistant to these targeted drugs just when they seem to be working perfectly. Now, two groups of researchers have developed rational combination treatments that block resistance to targeted cancer drugs by inhibiting mTOR. Elkabets and coauthors were working on breast cancer, where the PIK3CA gene is frequently mutated. Inhibitors of PI3K (the protein product of PIK3CA) are currently in clinical trials, but some of the cancers are resistant to these drugs. The authors have discovered that breast cancers resistant to the PI3K inhibitor BYL719 had persistently active mTOR signaling, both in cultured cell lines and in patient tumors. Adding an mTOR inhibitor to the treatment regimen could reverse the resistance and kill the tumor cells. Corcoran et al. found a similar mTOR-dependent drug resistance mechanism to be active in melanoma as well. BRAF-mutant melanomas, the most common type, are frequently treated with RAF and MEK inhibitors, but only with mixed results, because melanomas quickly develop resistance to these drugs. Now, the authors have shown that drug-resistant melanomas also have persistent activation of mTOR, and adding an mTOR inhibitor to the treatment regimen can block drug resistance and kill the cancer cells. In both studies, the activation of mTOR in drug-resistant tumors has been confirmed in human patients, but the combination treatments have only been tested in cells and in mouse models thus far. Thus, the next critical step would be to confirm that adding mTOR inhibition to treatment regimens for these cancers is effective in the clinical setting as well. Some mTOR inhibitors are already available for use in patients, so hopefully soon mTOR activation will not be something to beware of, but something to monitor and target with specific drugs. Activating mutations of the PIK3CA gene occur frequently in breast cancer, and inhibitors that are specific for phosphatidylinositol 3-kinase (PI3K) p110α, such as BYL719, are being investigated in clinical trials. In a search for correlates of sensitivity to p110α inhibition among PIK3CA-mutant breast cancer cell lines, we observed that sensitivity to BYL719 (as assessed by cell proliferation) was associated with full inhibition of signaling through the TORC1 pathway. Conversely, cancer cells that were resistant to BYL719 had persistently active mTORC1 signaling, although Akt phosphorylation was inhibited. Similarly, in patients, pS6 (residues 240/4) expression (a marker of mTORC1 signaling) was associated with tumor response to BYL719, and mTORC1 was found to be reactivated in tumors from patients whose disease progressed after treatment. In PIK3CA-mutant cancer cell lines with persistent mTORC1 signaling despite PI3K p110α blockade (that is, resistance), the addition of the allosteric mTORC1 inhibitor RAD001 to the cells along with BYL719 resulted in reversal of resistance in vitro and in vivo. Finally, we found that growth factors such as insulin-like growth factor 1 and neuregulin 1 can activate mammalian target of rapamycin (mTOR) and mediate resistance to BYL719. Our findings suggest that simultaneous administration of mTORC1 inhibitors may enhance the clinical activity of p110α-targeted drugs and delay the appearance of resistance.


Breast Cancer Research | 2008

NVP-AUY922: a small molecule HSP90 inhibitor with potent antitumor activity in preclinical breast cancer models

Michael Rugaard Jensen; Joseph Schoepfer; Thomas Radimerski; Andrew Massey; Chantale T. Guy; Josef Brueggen; Cornelia Quadt; Alan J. Buckler; Robert Cozens; Martin J. Drysdale; Carlos Garcia-Echeverria; Patrick Chène

IntroductionHeat shock protein 90 (HSP90) is a key component of a multichaperone complex involved in the post-translational folding of a large number of client proteins, many of which play essential roles in tumorigenesis. HSP90 has emerged in recent years as a promising new target for anticancer therapies.MethodsThe concentrations of the HSP90 inhibitor NVP-AUY922 required to reduce cell numbers by 50% (GI50 values) were established in a panel of breast cancer cell lines and patient-derived human breast tumors. To investigate the properties of the compound in vivo, the pharmacokinetic profile, antitumor effect, and dose regimen were established in a BT-474 breast cancer xenograft model. The effect on HSP90-p23 complexes, client protein degradation, and heat shock response was investigated in cell culture and breast cancer xenografts by immunohistochemistry, Western blot analysis, and immunoprecipitation.ResultsWe show that the novel small molecule HSP90 inhibitor NVP-AUY922 potently inhibits the proliferation of human breast cancer cell lines with GI50 values in the range of 3 to 126 nM. NVP-AUY922 induced proliferative inhibition concurrent with HSP70 upregulation and client protein depletion – hallmarks of HSP90 inhibition. Intravenous acute administration of NVP-AUY922 to athymic mice (30 mg/kg) bearing subcutaneous BT-474 breast tumors resulted in drug levels in excess of 1,000 times the cellular GI50 value for about 2 days. Significant growth inhibition and good tolerability were observed when the compound was administered once per week. Therapeutic effects were concordant with changes in pharmacodynamic markers, including HSP90-p23 dissociation, decreases in ERBB2 and P-AKT, and increased HSP70 protein levels.ConclusionNVP-AUY922 is a potent small molecule HSP90 inhibitor showing significant activity against breast cancer cells in cellular and in vivo settings. On the basis of its mechanism of action, preclinical activity profile, tolerability, and pharmaceutical properties, the compound recently has entered clinical phase I breast cancer trials.


The Journal of Nuclear Medicine | 2010

89Zr-Bevacizumab PET of Early Antiangiogenic Tumor Response to Treatment with HSP90 Inhibitor NVP-AUY922

Wouter B. Nagengast; Maarten A. de Korte; Thijs H. Oude Munnink; Hetty Timmer-Bosscha; Wifred F. den Dunnen; Harry Hollema; Johan R. de Jong; Michael Rugaard Jensen; Cornelia Quadt; Carlos Garcia-Echeverria; Guus A.M.S. van Dongen; Marjolijn N. Lub-de Hooge; Carolien P. Schröder; Elisabeth G.E. de Vries

Angiogenesis is a critical step in tumor development, in which vascular endothelial growth factor (VEGF) is a key growth aspect. Heat shock protein 90 (HSP90), a molecular chaperone, is essential for the activity of key proteins involved in VEGF transcription. Currently, no biomarkers to predict the effect of, or monitor, HSP90 inhibition therapy in individual patients exist. 89Zr-bevacizumab PET provides a noninvasive tool to monitor tumor VEGF levels. The aim of this study was to investigate 89Zr-bevacizumab PET for early antiangiogenic tumor response evaluation of treatment with the new HSP90 inhibitor NVP-AUY922. In xenografts of A2780 and its cisplatin-resistant CP70 human ovarian cancer subline, 89Zr-bevacizumab small-animal PET was performed before and after NVP-AUY922 treatment and verified with histologic response and ex vivo tumor VEGF levels. Compared with pretreatment values, 2 wk of NVP-AUY922 treatment decreased 89Zr-bevacizumab uptake by 44.4% (P = 0.0003) in A2780 xenografts, whereas tumor uptake was not affected in CP70 xenografts. The same pattern was observed in A2780 and CP70 tumor VEGF levels, measured with enzyme-linked immunosorbent assay, and mean vessel density after NVP-AUY922 treatment. These findings coincided with reduction in the proliferation rate, assessed by Ki67 staining, in A2780 tumor tissue only. Conclusion: 89Zr-bevacizumab PET was in line with the antiangiogenic response and direct antitumor effects after NVP-AUY922 treatment, supporting the specificity of 89Zr-bevacizumab PET as a sensitive technique to monitor the antiangiogenic response of HSP90 inhibition in vivo.


Clinical Cancer Research | 2013

First-in-Human Phase I Dose-Escalation Study of the HSP90 Inhibitor AUY922 in Patients with Advanced Solid Tumors

C. Sessa; Geoffrey I. Shapiro; Kapil N. Bhalla; Carolyn D. Britten; Karen S. Jacks; Monica M. Mita; Vassiliki Papadimitrakopoulou; Tim Pluard; Thomas A. Samuel; Mikhail Akimov; Cornelia Quadt; Cristina Fernandez-Ibarra; Hong Lu; Stuart Bailey; Sandra Chica; Udai Banerji

Purpose: A phase I study was conducted with the primary objective of determining the maximum tolerated dose (MTD) of AUY922 in patients with advanced solid tumors. Secondary objectives included characterization of the safety, pharmacokinetic, and pharmacodynamic profiles. Patients and Methods: Patients with advanced solid tumors received 1-hour i.v. infusions of AUY922 once a week in a 28-day cycle. An adaptive Bayesian logistic regression model that employed observed dose-limiting toxicities (DLT) in the first treatment cycle was used to guide dose-escalation decisions, with the established MTD to be used in phase II studies. Results: One hundred and one patients were enrolled and explored at doses in the range of 2 to 70 mg/m2. DLTs occurred in 8 patients (22–70 mg/m2) and included diarrhea, asthenia/fatigue, anorexia, atrial flutter, and visual symptoms. At 70 mg/m2, the AUY922 concentration achieved was consistent with active concentrations in a range of xenograft models. There was evidence of target inhibition in peripheral blood mononuclear cells (HSP70 induction) and tumor (client protein depletion and reduction of metabolic activity by 18F-FDG PET). The recommended phase II dose (RP2D) of 70 mg/m2 was proposed on the basis of toxicity and pharmacokinetic and pharmacodynamic profiles. Conclusions: At the RP2D of 70 mg/m2, AUY922 exhibited acceptable tolerability, and phase II single-agent and combination studies have been initiated in patients with HER2-positive breast, gastric, and non–small cell lung cancers. Clin Cancer Res; 19(13); 3671–80. ©2013 AACR.


OncoTargets and Therapy | 2016

PI3K inhibitors as new cancer therapeutics: implications for clinical trial design

Cristian Massacesi; Emmanuelle di Tomaso; Patrick Urban; Caroline Germa; Cornelia Quadt; Lucia Trandafir; Paola Aimone; Nathalie Fretault; Bharani Dharan; Ranjana Tavorath; Samit Hirawat

The PI3K–AKT–mTOR pathway is frequently activated in cancer. PI3K inhibitors, including the pan-PI3K inhibitor buparlisib (BKM120) and the PI3Kα-selective inhibitor alpelisib (BYL719), currently in clinical development by Novartis Oncology, may therefore be effective as anticancer agents. Early clinical studies with PI3K inhibitors have demonstrated preliminary antitumor activity and acceptable safety profiles. However, a number of unanswered questions regarding PI3K inhibition in cancer remain, including: what is the best approach for different tumor types, and which biomarkers will accurately identify the patient populations most likely to benefit from specific PI3K inhibitors? This review summarizes the strategies being employed by Novartis Oncology to help maximize the benefits of clinical studies with buparlisib and alpelisib, including stratification according to PI3K pathway activation status, selective enrollment/target enrichment (where patients with PI3K pathway-activated tumors are specifically recruited), nonselective enrollment with mandatory tissue collection, and enrollment of patients who have progressed on previous targeted agents, such as mTOR inhibitors or endocrine therapy. An overview of Novartis-sponsored and Novartis-supported trials that are utilizing these approaches in a range of cancer types, including breast cancer, head and neck squamous cell carcinoma, non-small cell lung carcinoma, lymphoma, and glioblastoma multiforme, is also described.


Leukemia | 2008

Signalling profile and antitumour activity of the novel Hsp90 inhibitor NVP-AUY922 in multiple myeloma.

Thorsten Stühmer; A Zöllinger; D Siegmund; Manik Chatterjee; E Grella; Stefan Knop; M Kortüm; C Unzicker; Michael Rugaard Jensen; Cornelia Quadt; Patrick Chène; Joseph Schoepfer; Carlos Garcia-Echeverria; H. Einsele; H Wajant; Ralf Bargou

We as well as others have recently shown that Hsp90 is overexpressed in multiple myeloma (MM) and critically contributes to tumour cell survival. Pharmacologic blockade of Hsp90 has consistently been found to induce MM cell death. However, most data have been obtained with MM cell lines whereas knowledge about the molecular effects of pharmacologic Hsp90 blockade in primary tumour cells is limited. Furthermore, these investigations have so far focused on geldanamycin derivatives. We analysed the biochemical effects of a novel diarylisoxazole-based Hsp90 inhibitor (NVP-AUY922) on signalling pathways and cell death in a large set of primary MM tumour samples and in MM cell lines. Treated cells displayed the molecular signature and pharmacodynamic properties for abrogation of Hsp90 function, such as downregulation of multiple survival pathways and strong upregulation of Hsp70. NVP-AUY922 treatment efficiently induced MM cell apoptosis and revealed both sensitive and resistant subgroups. Sensitivity was not correlated with TP53 mutation or Hsp70 induction levels and stromal cells from the bone marrow microenvironment were unable to abrogate NVP-AUY922-induced apoptosis of MM cells. Thus, NVP-AUY922 may be a promising drug for treatment of MM and clinical studies are warranted.


European Journal of Cancer | 2010

89Zr-trastuzumab PET visualises HER2 downregulation by the HSP90 inhibitor NVP-AUY922 in a human tumour xenograft

Thijs H. Oude Munnink; Maarten A. de Korte; Wouter B. Nagengast; Hetty Timmer-Bosscha; Carolina P. Schröder; Johan R. de Jong; Guus A.M.S. van Dongen; Michael Rugaard Jensen; Cornelia Quadt; Marjolijn N. Lub-de Hooge; Elisabeth G.E. de Vries

NVP-AUY922, a potent heat shock protein (HSP) 90 inhibitor, downregulates the expression of many oncogenic proteins, including the human epidermal growth factor receptor-2 (HER2). Because HER2 downregulation is a potential biomarker for early response to HSP90-targeted therapies, we used the (89)Zr-labelled HER2 antibody trastuzumab to quantify the alterations in HER2 expression after NVP-AUY922 treatment with HER2 positron emission tomography (PET) imaging. The HER2 overexpressing human SKOV-3 ovarian tumour cell line was used for in vitro experiments and as xenograft model in nude athymic mice. In vitro HER2 membrane expression was assessed by flow cytometry and a radio-immuno assay with (89)Zr-trastuzumab. For in vivo evaluation, mice received 50mg/kg NVP-AUY922 intraperitoneally every other day. (89)Zr-trastuzumab was injected intravenously 6d before NVP-AUY922 treatment and after 3 NVP-AUY922 doses. MicroPET imaging was performed at 24, 72 and 144h post tracer injection followed by ex-vivo biodistribution and immunohistochemical staining. After 24h NVP-AUY922 treatment HER2 membrane expression showed profound reduction with flow cytometry (80%) and radio-immuno assay (75%). PET tumour quantification, showed a mean reduction of 41% (p=0.0001) in (89)Zr-trastuzumab uptake at 144h post tracer injection after NVP-AUY922 treatment. PET results were confirmed by ex-vivo (89)Zr-trastuzumab biodistribution and HER2 immunohistochemical staining. NVP-AUY922 effectively downregulates HER2, which can be monitored and quantified in vivo non-invasively with (89)Zr-trastuzumab PET. This technique is currently under clinical evaluation and might serve as an early biomarker for HSP90 inhibition in HER2 positive metastatic breast cancer patients.


Cancer Research | 2012

Abstract CT-01: BYL719, a next generation PI3K alpha specific inhibitor: Preliminary safety, PK, and efficacy results from the first-in-human study

Dejan Juric; Jordi Rodon; Ana M. Gonzalez-Angulo; Howard A. Burris; Johanna C. Bendell; Jordan Berlin; Mark R. Middleton; Douglas Bootle; Markus Boehm; Antonin Schmitt; Nicolas Rouyrre; Cornelia Quadt; José Baselga

Introduction: Constitutive activation of the phosphatidylinositol-3-kinase (PI3K) signaling pathway is implicated in many human cancers. Until recently, drugs that specifically inhibit the alpha isoform of PI3K that is activated by alterations in the PIK3CA gene have been missing. BYL719 is the first oral PI3K inhibitor that strongly and selectively inhibits the PI3K alpha isoform of PI3K. Its biological activity correlates with inhibition of various downstream signaling components of the PI3K/Akt pathway and it inhibits the proliferation of breast cancer cell lines harboring PIK3CA mutations. In vivo, BYL719 shows statistically significant dose-dependent anti-tumor efficacy in PIK3CA mutant xenograft models in rodents. Methods: BYL719 entered clinical investigation in 2010 exclusively in patients (pts) with advanced solid malignancies carrying an alteration in the PIK3CA gene. In the dose escalation phase, dose selection is guided by a Bayesian regression model with overdose control. As of 21-Sep-2011, a total of 25 pts with a variety of solid tumors, such as colon and breast cancer, have been enrolled and treated at once daily oral doses ranging from 30mg to 450mg. Results: The safety profile of the compound is characterized by mostly on-target toxicity, such as hyperglycemia (33% of pts), which was found more frequently at higher doses and is largely reversible with BYL719 interruption and treatment with oral anti-diabetics. Other commonly reported toxicities include nausea (38%) and decreased appetite, diarrhea, and vomiting (each 29%). 2 DLTs of hyperglycemia and nausea, both CTCAE grade 3, were observed in 2 pts out of 5 treated at 450mg/d. In humans, BYL719 has a low clearance, a half-life of 8.5 h and its exposure increases dose proportionally between 30mg/d and 450mg/d, displaying a low inter-individual variability in Cmax and AUC. Exposure levels reached at clinical doses above 270 mg/d correspond to exposures causing tumor stasis or regression in preclinical PIK3CA dependent xenograft models. First signs of clinical efficacy of BYL719 include 1 confirmed partial response in a patient with ER+ breast cancer treated at 270mg/d. In addition, preliminary data suggest that significant PET responses (PMR) and/or tumor shrinkage were achieved in 8 out of 17 evaluated pts. Three pts had prolonged stable disease (≥7 months) at doses below 270mg/d and overall 8 patients have been on the study for at least 4 months. Upon determination of the MTD for the once daily dosing regimen, ∼45 pts with PIK3CA altered solid tumors will be enrolled into a safety expansion arm. Also, the PK and MTD for twice daily administration of BYL719 will be investigated. Conclusion: The preliminary clinical data available so far suggest BYL719 to be well tolerated, and showing signs of clinical activity, with manageable side effects and a predictable PK profile. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr CT-01. doi:1538-7445.AM2012-CT-01


Molecular Cancer Therapeutics | 2013

Inhibition of HSP90 with AUY922 Induces Synergy in HER2-Amplified Trastuzumab-Resistant Breast and Gastric Cancer

Zev A. Wainberg; Adrian Anghel; Amy Rogers; Amrita J. Desai; Ondrej Kalous; Dylan Conklin; Raul Ayala; Neil A. O'Brien; Cornelia Quadt; Mikhail Akimov; Dennis J. Slamon; Richard S. Finn

HSP90 enables the activation of many client proteins of which the most clinically validated is HER2. NVP-AUY922, a potent HSP90 inhibitor, is currently in phase II clinical trials. To explore its potential clinical use in HER2-amplified breast and gastric cancers, we evaluated the effect of AUY922 alone and in combination with trastuzumab in both trastuzumab-sensitive and -resistant models. A panel of 16 human gastric and 45 breast cancer cell lines, including 16 HER2-amplified (3 and 13, respectively) cells, was treated with AUY922 over various concentrations. In both breast and gastric cancer, we used cell lines and xenograft models with conditioned trastuzumab-resistance to investigate the efficacy of AUY922 alongside trastuzumab. Effects of this combination on downstream markers were analyzed via Western blot analysis. AUY922 exhibited potent antiproliferative activity in the low nanomolar range (<40 nmol/L) for 59 of 61 cell lines. In both histologies, HER2-amplified cells expressed greater sensitivity to AUY than HER2-negative cells. In conditioned trastuzumab-resistant models, AUY922 showed a synergistic effect with trastuzumab. In vitro, the combination induced greater decreases in HER2, a G2 cell-cycle arrest, and increased apoptosis. In a trastuzumab-resistant gastric cancer xenograft model, the combination of AUY922 and trastuzumab showed greater antitumor efficacy than either drug alone. These data suggest that AUY922 in combination with trastuzumab has unique efficacy in trastuzumab-resistant models. The combination of HSP90 inhibition and direct HER2 blockade represents a novel approach to the treatment of HER2-amplified cancers and clinical trials based on the above data are ongoing. Mol Cancer Ther; 12(4); 509–19. ©2013 AACR.

Collaboration


Dive into the Cornelia Quadt's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

José Baselga

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Howard A. Burris

Sarah Cannon Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jordi Rodon

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge