Network


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

Hotspot


Dive into the research topics where Vilmos Adleff is active.

Publication


Featured researches published by Vilmos Adleff.


Nature | 2015

The genomic landscape of response to EGFR blockade in colorectal cancer

Andrea Bertotti; Eniko Papp; Siân Jones; Vilmos Adleff; Valsamo Anagnostou; Barbara Lupo; Mark Sausen; Jillian Phallen; Carolyn Hruban; Collin Tokheim; Noushin Niknafs; Monica Nesselbush; Karli Lytle; Francesco Sassi; Francesca Cottino; Giorgia Migliardi; Eugenia Rosalinda Zanella; Dario Ribero; Nadia Russolillo; Alfredo Mellano; Andrea Muratore; Gianluca Paraluppi; Mauro Salizzoni; Silvia Marsoni; Michael Kragh; Johan Lantto; Andrea Cassingena; Qing Kay Li; Rachel Karchin; Robert B. Scharpf

Colorectal cancer is the third most common cancer worldwide, with 1.2 million patients diagnosed annually. In late-stage colorectal cancer, the most commonly used targeted therapies are the monoclonal antibodies cetuximab and panitumumab, which prevent epidermal growth factor receptor (EGFR) activation. Recent studies have identified alterations in KRAS and other genes as likely mechanisms of primary and secondary resistance to anti-EGFR antibody therapy. Despite these efforts, additional mechanisms of resistance to EGFR blockade are thought to be present in colorectal cancer and little is known about determinants of sensitivity to this therapy. To examine the effect of somatic genetic changes in colorectal cancer on response to anti-EGFR antibody therapy, here we perform complete exome sequence and copy number analyses of 129 patient-derived tumour grafts and targeted genomic analyses of 55 patient tumours, all of which were KRAS wild-type. We analysed the response of tumours to anti-EGFR antibody blockade in tumour graft models and in clinical settings and functionally linked therapeutic responses to mutational data. In addition to previously identified genes, we detected mutations in ERBB2, EGFR, FGFR1, PDGFRA, and MAP2K1 as potential mechanisms of primary resistance to this therapy. Novel alterations in the ectodomain of EGFR were identified in patients with acquired resistance to EGFR blockade. Amplifications and sequence changes in the tyrosine kinase receptor adaptor gene IRS2 were identified in tumours with increased sensitivity to anti-EGFR therapy. Therapeutic resistance to EGFR blockade could be overcome in tumour graft models through combinatorial therapies targeting actionable genes. These analyses provide a systematic approach to evaluating response to targeted therapies in human cancer, highlight new mechanisms of responsiveness to anti-EGFR therapies, and delineate new avenues for intervention in managing colorectal cancer.


Cancer Discovery | 2017

Evolution of Neoantigen Landscape during Immune Checkpoint Blockade in Non–Small Cell Lung Cancer

Valsamo Anagnostou; Kellie N. Smith; Patrick M. Forde; Noushin Niknafs; Rohit Bhattacharya; James White; Theresa Zhang; Vilmos Adleff; Jillian Phallen; Neha Wali; Carolyn Hruban; Violeta Beleva Guthrie; Kristen Rodgers; Jarushka Naidoo; Hyunseok Kang; William H. Sharfman; Christos S. Georgiades; Franco Verde; Peter B. Illei; Qing Kay Li; Edward Gabrielson; Malcolm V. Brock; Cynthia A. Zahnow; Stephen B. Baylin; Robert B. Scharpf; Julie R. Brahmer; Rachel Karchin; Drew M. Pardoll; Victor E. Velculescu

Immune checkpoint inhibitors have shown significant therapeutic responses against tumors containing increased mutation-associated neoantigen load. We have examined the evolving landscape of tumor neoantigens during the emergence of acquired resistance in patients with non-small cell lung cancer after initial response to immune checkpoint blockade with anti-PD-1 or anti-PD-1/anti-CTLA-4 antibodies. Analyses of matched pretreatment and resistant tumors identified genomic changes resulting in loss of 7 to 18 putative mutation-associated neoantigens in resistant clones. Peptides generated from the eliminated neoantigens elicited clonal T-cell expansion in autologous T-cell cultures, suggesting that they generated functional immune responses. Neoantigen loss occurred through elimination of tumor subclones or through deletion of chromosomal regions containing truncal alterations, and was associated with changes in T-cell receptor clonality. These analyses provide insight into the dynamics of mutational landscapes during immune checkpoint blockade and have implications for the development of immune therapies that target tumor neoantigens.Significance: Acquired resistance to immune checkpoint therapy is being recognized more commonly. This work demonstrates for the first time that acquired resistance to immune checkpoint blockade can arise in association with the evolving landscape of mutations, some of which encode tumor neoantigens recognizable by T cells. These observations imply that widening the breadth of neoantigen reactivity may mitigate the development of acquired resistance. Cancer Discov; 7(3); 264-76. ©2017 AACR.See related commentary by Yang, p. 250This article is highlighted in the In This Issue feature, p. 235.


Nature Communications | 2015

Clinical implications of genomic alterations in the tumour and circulation of pancreatic cancer patients

Mark Sausen; Jillian Phallen; Vilmos Adleff; Siân Jones; Rebecca J. Leary; Michael T. Barrett; Valsamo Anagnostou; Sonya Parpart-Li; Derek Murphy; Qing Kay Li; Carolyn Hruban; Rob Scharpf; James White; Peter J. O'Dwyer; Peter J. Allen; James R. Eshleman; Craig B. Thompson; David S. Klimstra; David C. Linehan; Anirban Maitra; Ralph H. Hruban; Luis A. Diaz; Daniel D. Von Hoff; Julia S. Johansen; Jeffrey A. Drebin; Victor E. Velculescu

Pancreatic adenocarcinoma has the worst mortality of any solid cancer. In this study, to evaluate the clinical implications of genomic alterations in this tumour type, we perform whole-exome analyses of 24 tumours, targeted genomic analyses of 77 tumours, and use non-invasive approaches to examine tumour-specific mutations in the circulation of these patients. These analyses reveal somatic mutations in chromatin-regulating genes MLL, MLL2, MLL3 and ARID1A in 20% of patients that are associated with improved survival. We observe alterations in genes with potential therapeutic utility in over a third of cases. Liquid biopsy analyses demonstrate that 43% of patients with localized disease have detectable circulating tumour DNA (ctDNA) at diagnosis. Detection of ctDNA after resection predicts clinical relapse and poor outcome, with recurrence by ctDNA detected 6.5 months earlier than with CT imaging. These observations provide genetic predictors of outcome in pancreatic cancer and have implications for new avenues of therapeutic intervention.


Science Translational Medicine | 2017

Direct detection of early-stage cancers using circulating tumor DNA

Jillian Phallen; Mark Sausen; Vilmos Adleff; Alessandro Leal; Carolyn Hruban; James White; Valsamo Anagnostou; Jacob Fiksel; Stephen Cristiano; Eniko Papp; Savannah Speir; Thomas Reinert; Mai-Britt Worm Ørntoft; Brian Woodward; Derek Murphy; Sonya Parpart-Li; David Riley; Monica Nesselbush; Naomi Sengamalay; Andrew Georgiadis; Qing Kay Li; Mogens Rørbæk Madsen; Frank Viborg Mortensen; Joost Huiskens; Cornelis J. A. Punt; Nicole C.T. van Grieken; Remond J.A. Fijneman; G. A. Meijer; Hatim Husain; Robert B. Scharpf

Noninvasive liquid biopsy analysis of circulating tumor DNA permits direct detection of early-stage cancers. Finding smaller needles in haystacks The detection and analysis of cell-free DNA in patients’ blood are becoming increasingly accepted in oncology. However, this approach has generally been applied for the monitoring of patients with existing tumors. It has not been useful for early diagnosis of cancer because of insufficient sensitivity to detect really small tumors that only shed minute quantities of DNA into the blood, as well as difficulties with identifying cancer-associated genetic changes without knowing what mutations are present in the primary tumor. A method developed by Phallen et al., called targeted error correction sequencing, addresses both of these limitations and demonstrates the feasibility of detecting circulating cell-free DNA from many early tumors, suggesting its potential use for cancer screening. Early detection and intervention are likely to be the most effective means for reducing morbidity and mortality of human cancer. However, development of methods for noninvasive detection of early-stage tumors has remained a challenge. We have developed an approach called targeted error correction sequencing (TEC-Seq) that allows ultrasensitive direct evaluation of sequence changes in circulating cell-free DNA using massively parallel sequencing. We have used this approach to examine 58 cancer-related genes encompassing 81 kb. Analysis of plasma from 44 healthy individuals identified genomic changes related to clonal hematopoiesis in 16% of asymptomatic individuals but no alterations in driver genes related to solid cancers. Evaluation of 200 patients with colorectal, breast, lung, or ovarian cancer detected somatic mutations in the plasma of 71, 59, 59, and 68%, respectively, of patients with stage I or II disease. Analyses of mutations in the circulation revealed high concordance with alterations in the tumors of these patients. In patients with resectable colorectal cancers, higher amounts of preoperative circulating tumor DNA were associated with disease recurrence and decreased overall survival. These analyses provide a broadly applicable approach for noninvasive detection of early-stage tumors that may be useful for screening and management of patients with cancer.


Clinical Cancer Research | 2017

The effect of preservative and temperature on the analysis of circulating tumor DNA

Sonya Parpart-Li; Bjarne Bartlett; Maria Popoli; Vilmos Adleff; Laura Tucker; Rebecca Steinberg; Andrew Georgiadis; Jillian Phallen; Julie R. Brahmer; Nilofer A Azad; Ilene Browner; D. Laheru; Victor E. Velculescu; Mark Sausen; Luis A. Diaz

Purpose: Analysis of genomic alterations in cell-free DNA (cfDNA) is evolving as an approach to detect, monitor, and genotype malignancies. Methods to separate the liquid from the cellular fraction of whole blood for circulating tumor DNA (ctDNA) analyses have been largely unstudied, although these may be a critical consideration for assay performance. Experimental Design: To evaluate the influence of blood processing on cfDNA and ctDNA quality and yield, we compared the cfDNA levels in serum with those in plasma. Given the limitations of serum for ctDNA analyses, we evaluated the effects of two plasma processing approaches, K2EDTA and Cell-Free DNA BCT (BCT) tubes, on cfDNA and ctDNA recovery. A total of 45 samples from nine patients with cancer were collected in both tube types. Once collected, blood was processed into plasma immediately or kept at room temperature and processed into plasma at 1, 3, 5, or 7 days. Results: As early as 24 hours after collection, plasma isolated from blood collected in K2EDTA tubes contained an elevated level of cfDNA that increased over time compared with BCT tubes where no significant increase in cfDNA levels was observed. When samples from an additional six patients with cancer, collected in the same manner, were stored at 4°C in K2EDTA tubes over the course of 3 days, total cfDNA and ctDNA levels were comparable between samples collected in BCT tubes. At day 3, there was a trend toward a decrease in ctDNA levels in both tubes that was more pronounced when measuring the mutant allele fraction for cases stored at 4°C in K2EDTA tubes. Conclusions: In summary, methods of blood processing have a strong influence on cfDNA and ctDNA levels and should be a consideration when evaluating ctDNA in peripheral circulation. Clin Cancer Res; 23(10); 2471–7. ©2016 AACR.


Nature Communications | 2017

High grade serous ovarian carcinomas originate in the fallopian tube

S. Intidhar Labidi-Galy; Eniko Papp; Dorothy Hallberg; Noushin Niknafs; Vilmos Adleff; Michaël Noë; Rohit Bhattacharya; Marian Novak; Siân Jones; Jillian Phallen; Carolyn Hruban; Michelle S. Hirsch; Douglas I. Lin; Lauren Schwartz; Cecile L. Maire; Jean-Christophe Tille; Michaela Bowden; A. Ayhan; Laura D. Wood; Robert B. Scharpf; Robert J. Kurman; Tian Li Wang; Ie Ming Shih; Rachel Karchin; Ronny Drapkin; Victor E. Velculescu

High-grade serous ovarian carcinoma (HGSOC) is the most frequent type of ovarian cancer and has a poor outcome. It has been proposed that fallopian tube cancers may be precursors of HGSOC but evolutionary evidence for this hypothesis has been limited. Here, we perform whole-exome sequence and copy number analyses of laser capture microdissected fallopian tube lesions (p53 signatures, serous tubal intraepithelial carcinomas (STICs), and fallopian tube carcinomas), ovarian cancers, and metastases from nine patients. The majority of tumor-specific alterations in ovarian cancers were present in STICs, including those affecting TP53, BRCA1, BRCA2 or PTEN. Evolutionary analyses reveal that p53 signatures and STICs are precursors of ovarian carcinoma and identify a window of 7 years between development of a STIC and initiation of ovarian carcinoma, with metastases following rapidly thereafter. Our results provide insights into the etiology of ovarian cancer and have implications for prevention, early detection and therapeutic intervention of this disease.It has previously been proposed that high-grade serous ovarian carcinoma (HGSOC) may originate from the fallopian tube. Here, the authors analyze genetic aberrances in fallopian tube lesions, ovarian cancers, and metastases from HGSOC patients and establish the evolutionary origins of HGSOC in the fallopian tube.


Clinical Cancer Research | 2017

Establishment of patient-derived tumor xenograft models of epithelial ovarian cancer for preclinical evaluation of novel therapeutics

Joyce Liu; Sangeetha Palakurthi; Qing Zeng; Shan Zhou; Elena Ivanova; Wei Huang; Ioannis K. Zervantonakis; Laura M. Selfors; Yiping Shen; Colin C. Pritchard; Mei Zheng; Vilmos Adleff; Eniko Papp; Huiying Piao; Marian Novak; Susan Fotheringham; Gerburg Wulf; Jessie M. English; Paul Kirschmeier; Victor E. Velculescu; Cloud P. Paweletz; Gordon B. Mills; David M. Livingston; Joan S. Brugge; Ursula A. Matulonis; Ronny Drapkin

Purpose: Ovarian cancer is the leading cause of death from gynecologic malignancy in the United States, with high rates of recurrence and eventual resistance to cytotoxic chemotherapy. Model systems that allow for accurate and reproducible target discovery and validation are needed to support further drug development in this disease. Experimental Design: Clinically annotated patient-derived xenograft (PDX) models were generated from tumor cells isolated from the ascites or pleural fluid of patients undergoing clinical procedures. Models were characterized by IHC and by molecular analyses. Each PDX was luciferized to allow for reproducible in vivo assessment of intraperitoneal tumor burden by bioluminescence imaging (BLI). Plasma assays for CA125 and human LINE-1 were developed as secondary tests of in vivo disease burden. Results: Fourteen clinically annotated and molecularly characterized luciferized ovarian PDX models were generated. Luciferized PDX models retain fidelity to both the nonluciferized PDX and the original patient tumor, as demonstrated by IHC, array CGH, and targeted and whole-exome sequencing analyses. Models demonstrated diversity in specific genetic alterations and activation of PI3K signaling pathway members. Response of luciferized PDX models to standard-of-care therapy could be reproducibly monitored by BLI or plasma markers. Conclusions: We describe the establishment of a collection of 14 clinically annotated and molecularly characterized luciferized ovarian PDX models in which orthotopic tumor burden in the intraperitoneal space can be followed by standard and reproducible methods. This collection is well suited as a platform for proof-of-concept efficacy and biomarker studies and for validation of novel therapeutic strategies in ovarian cancer. Clin Cancer Res; 23(5); 1263–73. ©2016 AACR.


Cancer Research | 2016

Abstract 3957: Optimized plasma collection procedures for liquid biopsy analyses in cancer

Sonya Parpart-Li; Bjarne Bartlett; Maria Popoli; Vilmos Adleff; Julie R. Brahmer; Nilo Azad; Sarah Bonerigo; Ilene S. Browner; Amy Ryan; Victor E. Velculescu; Mark Sausen; Luis A. Diaz

Analyses of genomic alterations in cell-free DNA (cfDNA) shed from tumors into the blood stream is rapidly advancing as a method to detect and genotype cancer. Liquid biopsy analyses are a favorable alternative to invasive tissue approaches, particularly for cancers in organs where tissue is difficult to obtain. As these methods become a clinical standard, the method for blood collection and plasma isolation will be an important consideration to ensure optimal assay performance with limited degradation of circulating tumor DNA (ctDNA). We compared blood collection and plasma isolation using both K2EDTA and cell-free DNA Streck™ BCT tubes to determine the stability of cfDNA and ctDNA in blood at room temperature or at 4°C. Blood was collected from nine cancer patients with KRAS mutations using K2EDTA and Streck™ BCT tubes. Over the course of seven days, cell lysis occurred in K2EDTA tubes at room temperature, releasing germline DNA into the blood (1300% increase over seven days), which significantly increased the number of total genomic equivalents. Additionally, we found that the volume of plasma collected from both tube types during blood fractionation decreased over time when tubes were at room temperature (an average of 3.5 mL vs. 1.0 mL, respectively). To evaluate the effect of storage conditions on performance of each tube type, blood was also collected from six cancer patients with KRAS or EGFR mutations and either maintained at room temperature or stored at 4°C for up to three days. Taken together, K2EDTA tubes stored at 4°C prevented cell lysis and preserved the ctDNA in a manner equivalent to Streck™ BCT tubes. These data demonstrate that liquid biopsies can be collected using either tube type with similar performance and appropriate consideration of storage conditions. Citation Format: Sonya T. Parpart-Li, Bjarne Bartlett, Maria Popoli, Vilmos Adleff, Julie Brahmer, Nilo Azad, Sarah Bonerigo, Ilene Browner, Amy Ryan, Victor Velculescu, Mark Sausen, Luis A. Diaz. Optimized plasma collection procedures for liquid biopsy analyses in cancer. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 3957.


Cancer Research | 2015

Abstract 619: Identification of clinically actionable genomic alterations in the tumor and circulation of pancreatic cancer patients

Mark Sausen; Jillian Phallen; Vilmos Adleff; Siân Jones; Rebecca J. Leary; Karli Lytle; Sonya Parpart-Li; Derek Murphy; Michael T. Barrett; David C. Linehan; Anirban Maitra; Ralph H. Hruban; Daniel D. Von Hoff; Julia S. Johansen; Luis A. Diaz; Jeffrey A. Drebin; Victor E. Velculescu

Proceedings: AACR 106th Annual Meeting 2015; April 18-22, 2015; Philadelphia, PA Pancreatic adenocarcinoma has the worst overall mortality of any solid tumor, with only 6% of patients surviving after 5 years. To evaluate the clinical implications of genomic alterations in this low cellularity tumor type, we deeply sequenced the genomes of 134 enriched pancreatic adenocarcinomas from patients who underwent potentially curative resections. Given the low neoplastic cellularity of pancreatic cancers, we enriched for neoplastic cells either by macrodissection of primary tumors or by flow-sorting of tumor nuclei, and performed deep sequencing (high coverage) of these enriched samples using next-generation sequencing approaches. We obtained a total of >1Tb of sequence data, resulting in an average coverage within the target regions of >200-fold for each tumor analyzed by whole-exome sequencing and >750-fold for each tumor analyzed by targeted cancer gene sequencing. These approaches allowed us to identify sequence changes, including single base and small insertion or deletion mutations, as well as copy number alterations in >20,000 genes in the whole-exome analyses and in 116 specific genes in the targeted analyses. These analyses revealed that somatic mutation of chromatin remodeling genes were associated with improved progression-free and overall survival. Alterations in genes with potential clinical utility were observed in a majority of cases and included alterations of AKT1, AKT2, BRCA2, ERBB2, KIT, and PIK3CA. Non-invasive liquid biopsy analyses were performed before and after surgery to evaluate the presence of circulating tumor DNA in the plasma of 83 patients. Through these approaches, we were able to diagnose early stage pancreatic tumors in the majority of patients and to detect the presence of circulating tumor DNA prior to clinical relapse. These observations provide genetic markers of clinical outcome in pancreatic cancer and suggest new avenues for personalized therapy. Citation Format: Mark Sausen, Jillian Phallen, Vilmos Adleff, Siân Jones, Rebecca J. Leary, Karli Lytle, Sonya Parpart-Li, Derek Murphy, Michael T. Barrett, David C. Linehan, Anirban Maitra, Ralph Hruban, Daniel D. Von Hoff, Julia S. Johansen, Luis A. Diaz, Jeffrey A. Drebin, Victor E. Velculescu. Identification of clinically actionable genomic alterations in the tumor and circulation of pancreatic cancer patients. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 619. doi:10.1158/1538-7445.AM2015-619


Journal of Clinical Oncology | 2018

Circulating tumor DNA dynamics in resectable gastric cancer.

Alessandro Leal; Nicole C.T. van Grieken; Jillian Phallen; Doreen N. Palsgrove; Vilmos Adleff; Fabienne Warmerdam; Donald L. van der Peet; Henk M.W. Verheul; D.J. van Spronsen; Johanna Mg Van Riel; Maud M. Geenen; Johanna Elisabeth A. Portielje; Hanneke W. M. van Laarhoven; Edwin P.M. Jansen; Karolina Sikorska; Elma Klein Kranenbarg; Cornelis J. H. van de Velde; Marcel Verheij; Annemieke Cats; Victor E. Velculescu

Collaboration


Dive into the Vilmos Adleff's collaboration.

Top Co-Authors

Avatar

Jillian Phallen

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Valsamo Anagnostou

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Mark Sausen

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Carolyn Hruban

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Luis A. Diaz

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Eniko Papp

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Alessandro Leal

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Rachel Karchin

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge