Network


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

Hotspot


Dive into the research topics where Costanza Paoletti is active.

Publication


Featured researches published by Costanza Paoletti.


Nature Nanotechnology | 2013

Sensitive capture of circulating tumour cells by functionalized graphene oxide nanosheets

Hyeun Joong Yoon; Tae Hyun Kim; Zhuo Zhang; Ebrahim Azizi; Trinh Pham; Costanza Paoletti; Jules Lin; Nithya Ramnath; Max S. Wicha; Daniel F. Hayes; Diane M. Simeone; Sunitha Nagrath

The spread of cancer throughout the body is driven by circulating tumour cells (CTCs)1. These cells detach from the primary tumour and move from the blood stream to a new site of subsequent tumour growth. They also carry information about the primary tumour and have the potential to be valuable biomarkers for disease diagnosis and progression, and for the molecular characterization of certain biological properties of the tumour. However, the limited sensitivity and specificity of current methods to measure and study these cells in patient blood samples prevent the realization of their full clinical potential. The use of microfluidic devices is a promising method for isolating CTCs2, 3; however, the devices are reliant on three-dimensional structures, which limit further characterization and expansion of cells on the chip. Here we demonstrate an effective approach to isolate CTCs from blood samples of pancreatic, breast and lung cancer patients, by using functionalised graphene oxide nanosheets on a patterned gold surface. CTCs were captured with high sensitivity at low concentration of target cells (73% ± 32.4 at 3–5 cells/mL blood).


Cancer and Metastasis Reviews | 2013

Clinical application of circulating tumor cells in breast cancer: overview of the current interventional trials

François-Clément Bidard; Tanja Fehm; Michail Ignatiadis; Jeffrey B. Smerage; Catherine Alix-Panabières; Wolfgang Janni; Carlo Messina; Costanza Paoletti; Volkmar Müller; Daniel F. Hayes; Martine Piccart; Jean-Yves Pierga

In 2004, circulating tumor cells (CTC) enumeration by the CellSearch® technique at baseline and during treatment was reported to be associated with prognosis in metastatic breast cancer patients. In 2008, the first evidence of the impact of CTC detection by this technique on survival of cM0(i+) patients were reported. These findings were confirmed by other non-interventional studies, whereas CTC were also investigated as a surrogate for tumor biology, mainly for HER2 expression/amplification. The aim of this report is to present the current prospective large interventional studies that have been specifically designed to demonstrate that CTC enumeration/characterization may improve the management of breast cancer patients: STIC CTC METABREAST (France) and Endocrine Therapy Index (USA) assess the CTC-guided hormone therapy vs chemotherapy decision in M1 patients; SWOG0500 (USA) and CirCe01 (France) assess the CTC count changes during treatment in metastatic patients; DETECT III (M1 patients, Germany) and Treat CTC (cM0(i+) patients, European Organization for Research and Treatment of Cancer/Breast International Group) assess the use of anti-HER2 treatments in HER2-negative breast cancer patients selected on the basis of CTC detection/characterization. These trials have different designs in various patient populations but are expected to be the pivotal trials for CTC implementation in the routine management of breast cancer patients.


Clinical Cancer Research | 2016

ESR1 mutations in circulating plasma tumor DNA from metastatic breast cancer patients

David Chu; Costanza Paoletti; Christina Gersch; Dustin A. VanDenBerg; Daniel J. Zabransky; Rory L. Cochran; Hong Yuen Wong; Patricia Valda Toro; Justin Cidado; Sarah Croessmann; Bracha Erlanger; Karen Cravero; Kelly Kyker-Snowman; Berry Button; Heather A. Parsons; W. Brian Dalton; Riaz Gillani; Arielle Medford; Kimberly Aung; Nahomi Tokudome; Arul M. Chinnaiyan; Anne F. Schott; Dan R. Robinson; Karen S. Jacks; Josh Lauring; Paula J. Hurley; Daniel F. Hayes; James M. Rae; Ben Ho Park

Purpose: Mutations in the estrogen receptor (ER)α gene, ESR1, have been identified in breast cancer metastases after progression on endocrine therapies. Because of limitations of metastatic biopsies, the reported frequency of ESR1 mutations may be underestimated. Here, we show a high frequency of ESR1 mutations using circulating plasma tumor DNA (ptDNA) from patients with metastatic breast cancer. Experimental Design: We retrospectively obtained plasma samples from eight patients with known ESR1 mutations and three patients with wild-type ESR1 identified by next-generation sequencing (NGS) of biopsied metastatic tissues. Three common ESR1 mutations were queried for using droplet digital PCR (ddPCR). In a prospective cohort, metastatic tissue and plasma were collected contemporaneously from eight ER-positive and four ER-negative patients. Tissue biopsies were sequenced by NGS, and ptDNA ESR1 mutations were analyzed by ddPCR. Results: In the retrospective cohort, all corresponding mutations were detected in ptDNA, with two patients harboring additional ESR1 mutations not present in their metastatic tissues. In the prospective cohort, three ER-positive patients did not have adequate tissue for NGS, and no ESR1 mutations were identified in tissue biopsies from the other nine patients. In contrast, ddPCR detected seven ptDNA ESR1 mutations in 6 of 12 patients (50%). Conclusions: We show that ESR1 mutations can occur at a high frequency and suggest that blood can be used to identify additional mutations not found by sequencing of a single metastatic lesion. Clin Cancer Res; 22(4); 993–9. ©2015 AACR.


Clinical Cancer Research | 2015

Development of Circulating Tumor Cell-Endocrine Therapy Index in Patients with Hormone Receptor Positive Breast Cancer

Costanza Paoletti; Maria C. Muñiz; Dafydd G. Thomas; Kent A. Griffith; Kelley M. Kidwell; Nahomi Tokudome; Martha E. Brown; Kimberly Aung; M. Craig Miller; Dorothy L. Blossom; Anne F. Schott; N. Lynn Henry; James M. Rae; Mark Carle Connelly; David Chianese; Daniel F. Hayes

Background: Endocrine therapy (ET) fails to induce a response in one half of patients with hormone receptor (HR)–positive metastatic breast cancer (MBC), and almost all will eventually become refractory to ET. Circulating tumor cells (CTC) are associated with worse prognosis in patients with MBC, but enumeration alone is insufficient to predict the absolute odds of benefit from any therapy, including ET. We developed a multiparameter CTC-Endocrine Therapy Index (CTC-ETI), which we hypothesize may predict resistance to ET in patients with HR-positive MBC. Methods: The CTC-ETI combines enumeration and CTC expression of four markers: estrogen receptor (ER), B-cell lymphoma 2 (BCL-2), Human Epidermal Growth Factor Receptor 2 (HER2), and Ki67. The CellSearch System and reagents were used to capture CTC and measure protein expression by immunofluorescent staining on CTC. Results: The feasibility of determining CTC-ETI was initially established in vitro and then in a prospective single-institution pilot study in patients with MBC. CTC-ETI was successfully determined in 44 of 50 (88%) patients. Eighteen (41%), 9 (20%), and 17 (39%) patients had low, intermediate, and high CTC-ETI scores, respectively. Interobserver concordance of CTC-ETI determination was from 94% to 95% (Kappa statistic, 0.90–0.91). Inter- and cell-to-cell intrapatient heterogeneity of expression of each of the CTC markers was observed. CTC biomarker expression was discordant from both primary and metastatic tissues. Conclusions: CTC expression of ER, BCL-2, HER2, and Ki67 can be reproducibly measured with high analytical validity using the CellSearch System. The clinical implications of CTC-ETI, and of the heterogeneity of CTC biomarker expression, are being evaluated in an ongoing prospective trial. Clin Cancer Res; 21(11); 2487–98. ©2014 AACR. See related commentary by Mathew et al., p. 2421


Clinical Biochemistry | 2016

Comparative analysis of circulating tumor DNA stability In K3EDTA, Streck, and CellSave blood collection tubes.

Qing Kang; N. Lynn Henry; Costanza Paoletti; Hui Jiang; Pankaj Vats; Arul M. Chinnaiyan; Daniel F. Hayes; Sofia D. Merajver; James M. Rae; Muneesh Tewari

OBJECTIVES Optimal conditions for blood collection for circulating tumor DNA (ctDNA) are still being developed. Although both Streck and EDTA tubes are commonly used, their ability to stabilize ctDNA as a function of time and temperature post-collection has not been thoroughly studied. Additionally, the potential utility of CellSave tubes (commonly used for circulating tumor cell) for ctDNA measurements has not been studied. DESIGN AND METHODS Blood was collected into Streck, EDTA, and CellSave tubes from ten patients with metastatic breast cancer enrolled in the MI-ONCOSEQ tumor sequencing program at the University of Michigan and kept either on ice or at room temperature until plasma isolation. Plasma was processed after 2, 6, and 48h post-collection. We used droplet digital PCR (ddPCR) to quantify plasma ctDNA and wild-type DNA for six patients who had tumor tissue mutations represented in commercially available ddPCR assays. RESULTS ctDNA abundance was similar and stable for up to 6h in all tube types, and there was no effect of storage temperature on the yield for Streck and EDTA tubes. After 48h, however, one out of four patients with detectable ctDNA showed a ~50% decline in ctDNA in the EDTA tube, and three out of six patients showed a 2-3-fold increase in wild-type DNA in the EDTA tube. CONCLUSIONS Streck, EDTA, and CellSave tubes showed similar performance in preserving ctDNA for up to 6h before plasma isolation. Streck and CellSave tubes more consistently stabilized ctDNA and wild-type DNA at 48h than EDTA tubes.


Journal of Internal Medicine | 2013

Circulating tumour cells: Insights into tumour heterogeneity

Daniel F. Hayes; Costanza Paoletti

Tumour heterogeneity is a major barrier to cure breast cancer. It can exist between patients with different intrinsic subtypes of breast cancer or within an individual patient with breast cancer. In the latter case, heterogeneity has been observed between different metastatic sites, between metastatic sites and the original primary tumour, and even within a single tumour at either a metastatic or a primary site. Tumour heterogeneity is a function of two separate, although linked, processes. First, genetic instability is a hallmark of malignancy, and results in ‘fixed’ genetic changes that are almost certainly carried forward through progression of the cancer over time, with increasingly complex additional genetic changes in new metastases as they arise. The second type of heterogeneity is due to differential but ‘plastic’ expression of various genes important in the biology and response to various therapies. Together, these processes result in highly variable cancers with differential response, and resistance, to both targeted (e.g. endocrine or anti‐human epithelial growth receptor type 2 (HER2) agents) and nontargeted therapies (e.g. chemotherapy). Ideally, tumour heterogeneity would be monitored over time, especially in relation to therapeutic strategies. However, biopsies of metastases require invasive and costly procedures, and biopsies of multiple metastases, or serially over time, are impractical. Circulating tumour cells (CTCs) represent a potential surrogate for tissue‐based cancer and therefore might provide the opportunity to monitor serial changes in tumour biology. Recent advances have enabled accurate and reliable quantification and molecular characterization of CTCs with regard to a number of important biomarkers including oestrogen receptor alpha and HER2. Preliminary data have demonstrated that expression of these markers between CTCs in individual patients with metastatic breast cancer reflects the heterogeneity of the underlying tumours. Future studies are designed to determine the clinical utility of these novel technologies in either research or routine clinical settings.


Annual Review of Medicine | 2014

Molecular Testing in Breast Cancer

Costanza Paoletti; Daniel F. Hayes

Tumor biomarker tests are critical to implementation of personalized medicine for patients at risk for or affected by breast cancer. A tumor biomarker test must have high analytical validity and clinical utility to be used to guide clinical care in standard practice. Few tumor biomarkers meet these high standards. These include germline DNA single-nucleotide polymorphisms in the BRCA1 and -2 genes to determine high risk in unaffected women, selected tissue-based markers to determine prognosis and predict benefit from therapy, and circulating MUC1, CEA and perhaps tumor cells to monitor patients with metastatic disease. Efforts to discover biomarkers that predict therapeutic toxicity are promising but not yet successful. Further research is needed to enhance the number of tumor biomarker tests so that patients with breast cancer can get the correct treatment at the appropriate time.


Molecular Oncology | 2016

Heterogeneous estrogen receptor expression in circulating tumor cells suggests diverse mechanisms of fulvestrant resistance

Costanza Paoletti; Jose M. Larios; Maria C. Muñiz; Kimberly Aung; Emily M. Cannell; Elizabeth P. Darga; Kelley M. Kidwell; Dafydd G. Thomas; Nahomi Tokudome; Martha E. Brown; Mark C. Connelly; David A. Chianese; Anne F. Schott; N. Lynn Henry; James M. Rae; Daniel F. Hayes

Fulvestrant is a dose dependent selective estrogen receptor (ER) down‐regulator (SERD) used in ER‐positive metastatic breast cancer (MBC). Nearly all patients develop resistance. We performed molecular analysis of circulating tumor cells (CTC) to gain insight into fulvestrant resistance.


Cancer Research | 2013

Abstract OT1-3-01: Characterization of circulating tumor cells from subjects with metastatic breast cancer using the CTC-endocrine therapy index: The COMETI-P2-2012.0 trial

Costanza Paoletti

Introduction: Approximately 50% of patients with estrogen receptor (ER) positive metastatic breast cancer (MBC) benefit from endocrine therapy (ET). This percentage is even lower after first line ET. Other than absence of ER, there are no validated markers that predict lack of benefit from ET. Prior studies suggest that relative levels of four markers might predict lack of benefit from ET: low ER and BCL2 and high HER2, and Ki67. Elevated circulating tumor cell (CTC) levels are prognostic in patients with MBC. We have recently demonstrated the ability to quantify ER, BCL2, HER2 and Ki67 on CTC using the CellSearch® system, and we have developed an analytically validated CTC-Endocrine Therapy Index (CTC-ETI). We are now conducting a prospective, multi-institutional clinical trial to test whether pretreatment CTC-ETI predicts lack of benefit from ET, and whether serial CTC-ETI measurements can be used to monitor patients with ER positive MBC starting 2nd or later line of ET. Objectives: To demonstrate that CTC-ETI can be accurately determined in multiple centers across North America and whether high CTC-ETI is associated with rapid progression (within 3 months of starting ET), and finally to refine the current CTC-ETI algorithm. Secondary objectives: To determine if changes in CTC-ETI during ET are associated with rapid progression, to correlate the status of the biomarkers on baseline CTC with the status of the same biomarkers in primary and/or metastatic tissue collected from the subjects, and to store CTC positive cartridges for future molecular analyses. Methods: Women with ER positive, HER2 negative, progressive MBC after one or more lines of ET or during or within 12 months of completing adjuvant ET who are initiating a new ET are eligible. Subjects with brain metastases only or those who are progressing on fulvestrant are not eligible. All subjects must provide written informed consent. Patients may be treated with any approved ET for MBC, and may also receive everolimus. Patients may not be on any investigational therapy. Blood draws (approximately 40 ml) are collected at baseline, at 1, 2, and 3 months after starting therapy, and at 12 months or progression. CTC-ETI is performed as previously described. Statistical Plan: Enrollment of 200 subjects will provide a sufficient number of evaluable subjects and statistical power for evaluation of the primary and secondary objectives. A total of 120 subjects evaluable for clinical validity analyses will be required to obtain the minimum of 51 subjects with rapid progression for evaluation of the clinical validity of the CTC-ETI. Subjects will be considered evaluable for clinical validity if they have successful calculation of a baseline CTC-ETI and progression status within 3 months of starting ET can be ascertained. Current Status: The first patient was enrolled in May, 2013, and accrual is ongoing. Sponsored by Veridex, LLC. Citation Information: Cancer Res 2013;73(24 Suppl): Abstract nr OT1-3-01.


Cancer Research | 2017

Comprehensive mutation and copy number profiling in archived circulating breast cancer tumor cells documents heterogeneous resistance mechanisms

Costanza Paoletti; Andi K. Cani; Jose M. Larios; Daniel H. Hovelson; Kimberly Aung; Elizabeth P. Darga; Emily M. Cannell; Paul J. Baratta; Chia Jen Liu; David Chu; Maryam Yazdani; Allen R. Blevins; Valeria Sero; Nahomi Tokudome; Dafydd G. Thomas; Christina Gersch; Anne F. Schott; Yi Mi Wu; Robert J. Lonigro; Dan R. Robinson; Arul M. Chinnaiyan; Farideh Z. Bischoff; Michael D. Johnson; Ben Ho Park; Daniel F. Hayes; James M. Rae; Scott A. Tomlins

Addressing drug resistance is a core challenge in cancer research, but the degree of heterogeneity in resistance mechanisms in cancer is unclear. In this study, we conducted next-generation sequencing (NGS) of circulating tumor cells (CTC) from patients with advanced cancer to assess mechanisms of resistance to targeted therapy and reveal opportunities for precision medicine. Comparison of the genomic landscapes of CTCs and tissue metastases is complicated by challenges in comprehensive CTC genomic profiling and paired tissue acquisition, particularly in patients who progress after targeted therapy. Thus, we assessed by NGS somatic mutations and copy number alterations (CNA) in archived CTCs isolated from patients with metastatic breast cancer who were enrolled in concurrent clinical trials that collected and analyzed CTCs and metastatic tissues. In 76 individual and pooled informative CTCs from 12 patients, we observed 85% concordance in at least one or more prioritized somatic mutations and CNA between paired CTCs and tissue metastases. Potentially actionable genomic alterations were identified in tissue but not CTCs, and vice versa. CTC profiling identified diverse intra- and interpatient molecular mechanisms of endocrine therapy resistance, including loss of heterozygosity in individual CTCs. For example, in one patient, we observed CTCs that were either wild type for ESR1 (n = 5/32), harbored the known activating ESR1 p.Y537S mutation (n = 26/32), or harbored a novel ESR1 p.A569S (n = 1/32). ESR1 p.A569S was modestly activating in vitro, consistent with its presence as a minority circulating subclone. Our results demonstrate the feasibility and potential clinical utility of comprehensive profiling of archived fixed CTCs. Tissue and CTC genomic assessment are complementary, and precise combination therapies will likely be required for effective targeting in advanced breast cancer patients.Significance: These findings demonstrate the complementary nature of genomic profiling from paired tissue metastasis and circulating tumor cells from patients with metastatic breast cancer. Cancer Res; 78(4); 1110-22. ©2017 AACR.

Collaboration


Dive into the Costanza Paoletti's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge