Network


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

Hotspot


Dive into the research topics where V.A. Miller is active.

Publication


Featured researches published by V.A. Miller.


Nature Medicine | 2013

Oncogenic and drug-sensitive NTRK1 rearrangements in lung cancer

Aria Vaishnavi; Marzia Capelletti; Anh T. Le; Severine Kako; Mohit Butaney; Dalia Ercan; Sakshi Mahale; Kurtis D. Davies; Dara L. Aisner; Amanda B. Pilling; Eamon M. Berge; Jhingook Kim; Hidefumi Sasaki; Seung-Il Park; Gregory V. Kryukov; Levi A. Garraway; Peter S. Hammerman; Julia Haas; Steven W. Andrews; Doron Lipson; Philip J. Stephens; V.A. Miller; Marileila Varella-Garcia; Pasi A. Jänne; Robert C. Doebele

We identified new gene fusions in patients with lung cancer harboring the kinase domain of the NTRK1 gene that encodes the high-affinity nerve growth factor receptor (TRKA protein). Both the MPRIP-NTRK1 and CD74-NTRK1 fusions lead to constitutive TRKA kinase activity and are oncogenic. Treatment of cells expressing NTRK1 fusions with inhibitors of TRKA kinase activity inhibited autophosphorylation of TRKA and cell growth. Tumor samples from 3 of 91 patients with lung cancer (3.3%) without known oncogenic alterations assayed by next-generation sequencing or fluorescence in situ hybridization demonstrated evidence of NTRK1 gene fusions.


Gynecologic Oncology | 2013

Comprehensive genomic profiling of epithelial ovarian cancer by next generation sequencing-based diagnostic assay reveals new routes to targeted therapies

J.S. Ross; Siraj M. Ali; Kai Wang; Gary A. Palmer; Roman Yelensky; Doron Lipson; V.A. Miller; D. Zajchowski; L.K. Shawver; P.J. Stephens

OBJECTIVE Targeted next generation sequencing (NGS) was evaluated for its ability to identify unanticipated targetable genomic alterations (GA) for patients with relapsed ovarian epithelial carcinoma (OC). METHODS DNA sequencing was performed for 3320 exons of 182 cancer-related genes and 37 introns of 14 genes frequently rearranged in cancer on indexed, adaptor ligated, hybridization-captured libraries using DNA isolated from FFPE sections from 48 histologically verified relapsed OC specimens. The original primary tumor was sequenced in 26 (54%) of the cases and recurrent/metastatic tumor site biopsies were sequenced in 22 (46%) of the cases. Actionability was defined as: GA that predict sensitivity or resistance to approved or standard therapies or are inclusion or exclusion criteria for specific experimental therapies in NCI registered clinical trials. RESULTS There were 38 (80%) serous, 5 (10%) endometrioid, 3 (6%) clear cell, 1 mucinous (2%) and 1 (2%) undifferentiated carcinomas. 141 GA were identified with an average of 2.9 GA (range 0-8) per tumor, of which 67 were actionable for an average of 1.4 actionable GA per patient (range 0-5). 33/48 (69%) of OC patient samples harbored at least one actionable GA. Most common GA were TP53 (79%); MYC (25%); BRCA1/2 (23%); KRAS (16.6%) and NF1 (14.5%). One tumor featured an ERBB2 point mutation. One of 3 (33%) of clear cell tumors featured cMET amplification validated by both FISH and IHC. CONCLUSIONS NGS assessment of therapy resistant OC identifies an unexpectedly high frequency of GA that could influence targeted therapy selection for the disease.


Journal of Clinical Pathology | 2014

Next-generation sequencing reveals frequent consistent genomic alterations in small cell undifferentiated lung cancer.

J.S. Ross; Kai Wang; O R Elkadi; Ashley J. Tarasen; L Foulke; Christine E. Sheehan; Geoff Otto; Gary A. Palmer; Roman Yelensky; Doron Lipson; Juliann Chmielecki; Siraj M. Ali; Julia A. Elvin; Deborah Morosini; V.A. Miller; P.J. Stephens

Aims Small cell lung cancer (SCLC) carries a poor prognosis, and the systemic therapies currently used as treatments are only modestly effective, as demonstrated by a low 5-year survival at only ∼5%. In this retrospective collected from March 2013 to study, we performed comprehensive genomic profiling of 98 small cell undifferentiated lung cancer (SCLC) samples to identify potential targets of therapy not currently searched for in routine clinical practice. Methods DNA from 98 SCLC was sequenced to high, uniform coverage (Illumina HiSeq 2500) and analysed for all classes of genomic alterations. Results A total of 386 alterations were identified for an average of 3.9 alterations per tumour (range 1–10). Fifty-two (53%) of cases harboured at least 1 actionable alteration with the potential to personalise therapy including base substitutions, amplifications or homozygous deletions in RICTOR (10%), KIT (7%), PIK3CA (6%), EGFR (5%), PTEN (5%), KRAS (5%), MCL1 (4%), FGFR1 (4%), BRCA2, (4%), TSC1 (3%), NF1 (3%), EPHA3 (3%) and CCND1. The most common non-actionable genomic alterations were alterations in TP53 (86% of SCLC cases), RB1 (54%) and MLL2 (17%). Conclusions Greater than 50% of the SCLC cases harboured at least one actionable alteration. Given the limited treatment options and poor prognosis of patients with SCLC, comprehensive genomic profiling has the potential to identify new treatment paradigms and meet an unmet clinical need for this disease.


Annals of Oncology | 2016

Comprehensive genomic profiling of anal squamous cell carcinoma reveals distinct genomically defined classes

Jon Chung; Eric M. Sanford; Adrienne Johnson; Samuel J. Klempner; Alexa B. Schrock; Norma Alonzo Palma; Rachel L. Erlich; Garrett Michael Frampton; Zachary R. Chalmers; Jo-Anne Vergilio; Douglas A. Rubinson; James Sun; Juliann Chmielecki; Roman Yelensky; James Suh; Doron Lipson; Thomas J. George; Julia A. Elvin; P.J. Stephens; V.A. Miller; J.S. Ross; Siraj M. Ali

BACKGROUND Squamous cell cancers of the anal canal (ASCC) are increasing in frequency and lack effective therapies for advanced disease. Although an association with human papillomavirus (HPV) has been established, little is known about the molecular characterization of ASCC. A comprehensive genomic analysis of ASCC was undertaken to identify novel genomic alterations (GAs) that will inform therapeutic choices for patients with advanced disease. PATIENTS AND METHODS Hybrid-capture-based next-generation sequencing of exons from 236 cancer-related genes and intronic regions from 19 genes commonly rearranged in cancer was performed on 70 patients with ASCC. HPV status was assessed by aligning tumor sequencing reads to HPV viral genomes. GAs were identified using an established algorithm and correlated with HPV status. RESULTS Sixty-one samples (87%) were HPV-positive. A mean of 3.5 GAs per sample was identified. Recurrent alterations in phosphoinositol-3-kinase pathway (PI3K/AKT/mTOR) genes including amplifications and homozygous deletions were present in 63% of cases. Clinically relevant GAs in genes involved in DNA repair, chromatin remodeling, or receptor tyrosine kinase signaling were observed in 30% of cases. Loss-of-function mutations in TP53 and CDKN2A were significantly enhanced in HPV-negative cases (P < 0.0001). CONCLUSIONS This is the first comprehensive genomic analysis of ASCC, and the results suggest new therapeutic approaches. Differing genomic profiles between HPV-associated and HPV-negative ASCC warrants further investigation and may require novel therapeutic and preventive strategies.


Journal of Clinical Pathology | 2014

Next-generation sequencing of adrenocortical carcinoma reveals new routes to targeted therapies

J.S. Ross; Kai Wang; J V Rand; M J Presta; Christine E. Sheehan; Siraj M. Ali; Julia A. Elvin; Elaine LaBrecque; C Hiemstra; Jamie Buell; Geoff Otto; Roman Yelensky; Doron Lipson; Deborah Morosini; Juliann Chmielecki; V.A. Miller; P.J. Stephens

Aims Adrenocortical carcinoma (ACC) carries a poor prognosis and current systemic cytotoxic therapies result in only modest improvement in overall survival. In this retrospective study, we performed a comprehensive genomic profiling of 29 consecutive ACC samples to identify potential targets of therapy not currently searched for in routine clinical practice. Methods DNA from 29 ACC was sequenced to high, uniform coverage (Illumina HiSeq) and analysed for genomic alterations (GAs). Results At least one GA was found in 22 (76%) ACC (mean 2.6 alterations per ACC). The most frequent GAs were in TP53 (34%), NF1 (14%), CDKN2A (14%), MEN1 (14%), CTNNB1 (10%) and ATM (10%). APC, CCND2, CDK4, DAXX, DNMT3A, KDM5C, LRP1B, MSH2 and RB1 were each altered in two cases (7%) and EGFR, ERBB4, KRAS, MDM2, NRAS, PDGFRB, PIK3CA, PTEN and PTCH1 were each altered in a single case (3%). In 17 (59%) of ACC, at least one GA was associated with an available therapeutic or a mechanism-based clinical trial. Conclusions Next-generation sequencing can discover targets of therapy for relapsed and metastatic ACC and shows promise to improve outcomes for this aggressive form of cancer.


Clinical Lymphoma, Myeloma & Leukemia | 2014

Vemurafenib Response in 2 Patients With Posttransplant Refractory BRAF V600E–Mutated Multiple Myeloma

Jeff P. Sharman; Juliann Chmielecki; Deborah Morosini; Gary A. Palmer; J.S. Ross; P.J. Stephens; J. Stafl; V.A. Miller; Siraj M. Ali

Multiple myeloma is commonly associated with genomic alterations that result in hyperactivation of the mitogen-activated protein kinase pathway. BRAF (v-raf murine sarcoma viral oncogene homolog B) V600E activating mutations have been observed in 4% of multiple myeloma cases. Patients with BRAF V600Eemutated myeloma may have an unusually aggressive clinical course associated with prominent extramedullary disease and a short duration of response to standard therapies. Vemurafenib, a BRAF V600Eespecific inhibitor and an FDA approved agent for treatment of melanoma, also has clinical activity in BRAF V600E mutationepositive multiple myeloma.


Leukemia | 2017

Patient-derived xenotransplants can recapitulate the genetic driver landscape of acute leukemias.

Kai Wang; M Sanchez-Martin; Xujun Wang; Kristina M. Knapp; Richard Koche; Ly P. Vu; Michelle Nahas; Jie He; Michael Hadler; Eytan M. Stein; Martin S. Tallman; Amy Donahue; Garrett Michael Frampton; Doron Lipson; Steven Roels; Phil Stephens; E M Sanford; Tim Brennan; Geoff Otto; Roman Yelensky; V.A. Miller; Michael G. Kharas; Ross L. Levine; Adolfo A. Ferrando; Scott A. Armstrong; Andrei V. Krivtsov

Genomic studies have identified recurrent somatic mutations in acute leukemias. However, current murine models do not sufficiently encompass the genomic complexity of human leukemias. To develop preclinical models, we transplanted 160 samples from patients with acute leukemia (acute myeloid leukemia, mixed lineage leukemia, B-cell acute lymphoblastic leukemia, T-cell ALL) into immunodeficient mice. Of these, 119 engrafted with expected immunophenotype. Targeted sequencing of 374 genes and 265 frequently rearranged RNAs detected recurrent and novel genetic lesions in 48 paired primary tumor (PT) and patient-derived xenotransplant (PDX) samples. Overall, the frequencies of 274 somatic variant alleles correlated between PT and PDX samples, although the data were highly variable for variant alleles present at 0–10%. Seventeen percent of variant alleles were detected in either PT or PDX samples only. Based on variant allele frequency changes, 24 PT-PDX pairs were classified as concordant while the other 24 pairs showed various degree of clonal discordance. There was no correlation of clonal concordance with clinical parameters of diseases. Significantly more bone marrow samples than peripheral blood samples engrafted discordantly. These data demonstrate the utility of developing PDX banks for modeling human leukemia, and emphasize the importance of genomic profiling of PDX and patient samples to ensure concordance before performing mechanistic or therapeutic studies.


Blood | 2015

Tumor-specific HSP90 inhibition as a therapeutic approach in JAK-mutant acute lymphoblastic leukemias.

Nicole Kucine; Sachie Marubayashi; Neha Bhagwat; Efthymia Papalexi; Priya Koppikar; Marta Sanchez Martin; Lauren Dong; Marty S. Tallman; Elisabeth Paietta; Kai Wang; Jie He; Doron Lipson; Phil Stephens; V.A. Miller; Jacob M. Rowe; Julie Teruya-Feldstein; Charles G. Mullighan; Adolfo A. Ferrando; Andrei V. Krivtsov; Scott A. Armstrong; Laura Leung; Stefan O. Ochiana; Gabriela Chiosis; Ross L. Levine; Maria Kleppe

The development of the dual Janus kinase 1/2 (JAK1/2) inhibitor ruxolitinib for the treatment of myeloproliferative neoplasms (MPNs) has led to studies of ruxolitinib in other clinical contexts, including JAK-mutated acute lymphoblastic leukemia (ALL). However, the limited ability of JAK inhibition to induce molecular or clinicopathological responses in MPNs suggests a need for development of better therapies for JAK kinase-dependent malignancies. Here, we demonstrate that heat shock protein 90 (HSP90) inhibition using a purine-scaffold HSP90 inhibitor in early clinical development is an effective therapeutic approach in JAK-dependent ALL and can overcome persistence to JAK-inhibitor therapy in ALL cells.


Annals of Oncology | 2017

Hybrid capture-based genomic profiling of circulating tumor DNA from patients with estrogen receptor-positive metastatic breast cancer.

Jon Chung; Dean Pavlick; Ryan J. Hartmaier; Alexa B. Schrock; Lauren Young; B Forcier; P Ye; M K Levin; Michael E. Goldberg; Howard A. Burris; A D Hoffman; P.J. Stephens; Garrett Michael Frampton; Doron Lipson; D M Nguyen; Shridar Ganesan; B H Park; Linda T. Vahdat; Brian Leyland-Jones; Tariq I. Mughal; Lajos Pusztai; J O’Shaughnessy; V.A. Miller; J.S. Ross; Siraj M. Ali

BACKGROUND Genomic changes that occur in breast cancer during the course of disease have been informed by sequencing of primary and metastatic tumor tissue. For patients with relapsed and metastatic disease, evolution of the breast cancer genome highlights the importance of using a recent sample for genomic profiling to guide clinical decision-making. Obtaining a metastatic tissue biopsy can be challenging, and analysis of circulating tumor DNA (ctDNA) from blood may provide a minimally invasive alternative. PATIENTS AND METHODS Hybrid capture-based genomic profiling was carried out on ctDNA from 254 female patients with estrogen receptor-positive breast cancer. Peripheral blood samples were submitted by clinicians in the course of routine clinical care between May 2016 and March 2017. Sequencing of 62 genes was carried out to a median unique coverage depth of 7503×. Genomic alterations (GAs) in ctDNA were evaluated and compared with matched tissue samples and genomic datasets of tissue from breast cancer. RESULTS At least 1GA was reported in 78% of samples. Frequently altered genes were TP53 (38%), ESR1 (31%) and PIK3CA (31%). Temporally matched ctDNA and tissue samples were available for 14 patients; 89% of mutations detected in tissue were also detected in ctDNA. Diverse ESR1 GAs including mutation, rearrangement and amplification, were observed. Multiple concurrent ESR1 GAs were observed in 40% of ESR1-altered cases, suggesting polyclonal origin; ESR1 compound mutations were also observed in two cases. ESR1-altered cases harbored co-occurring GAs in PIK3CA (35%), FGFR1 (16%), ERBB2 (8%), BRCA1/2 (5%), and AKT1 (4%). CONCLUSIONS GAs relevant to relapsed/metastatic breast cancer management were identified, including diverse ESR1 GAs. Genomic profiling of ctDNA demonstrated sensitive detection of mutations found in tissue. Detection of amplifications was associated with ctDNA fraction. Genomic profiling of ctDNA may provide a complementary and possibly alternative approach to tissue-based genomic testing for patients with estrogen receptor-positive metastatic breast cancer.Abstract Background Genomic changes that occur in breast cancer during the course of disease have been informed by sequencing of primary and metastatic tumor tissue. For patients with relapsed and metastatic disease, evolution of the breast cancer genome highlights the importance of using a recent sample for genomic profiling to guide clinical decision-making. Obtaining a metastatic tissue biopsy can be challenging, and analysis of circulating tumor DNA (ctDNA) from blood may provide a minimally invasive alternative. Patients and methods Hybrid capture-based genomic profiling was carried out on ctDNA from 254 female patients with estrogen receptor-positive breast cancer. Peripheral blood samples were submitted by clinicians in the course of routine clinical care between May 2016 and March 2017. Sequencing of 62 genes was carried out to a median unique coverage depth of 7503×. Genomic alterations (GAs) in ctDNA were evaluated and compared with matched tissue samples and genomic datasets of tissue from breast cancer. Results At least 1 GA was reported in 78% of samples. Frequently altered genes were TP53 (38%), ESR1 (31%) and PIK3CA (31%). Temporally matched ctDNA and tissue samples were available for 14 patients; 89% of mutations detected in tissue were also detected in ctDNA. Diverse ESR1 GAs including mutation, rearrangement and amplification, were observed. Multiple concurrent ESR1 GAs were observed in 40% of ESR1-altered cases, suggesting polyclonal origin; ESR1 compound mutations were also observed in two cases. ESR1-altered cases harbored co-occurring GAs in PIK3CA (35%), FGFR1 (16%), ERBB2 (8%), BRCA1/2 (5%), and AKT1 (4%). Conclusions GAs relevant to relapsed/metastatic breast cancer management were identified, including diverse ESR1 GAs. Genomic profiling of ctDNA demonstrated sensitive detection of mutations found in tissue. Detection of amplifications was associated with ctDNA fraction. Genomic profiling of ctDNA may provide a complementary and possibly alternative approach to tissue-based genomic testing for patients with estrogen receptor-positive metastatic breast cancer.


Lung Cancer | 2017

Emergence of EGFR G724S mutation in EGFR-mutant lung adenocarcinoma post progression on osimertinib

A. Oztan; S. Fischer; Alexa B. Schrock; Rachel L. Erlich; Christine M. Lovly; P.J. Stephens; Jeffrey S. Ross; V.A. Miller; Siraj M. Ali; S-H.I. Ou; Luis E. Raez

Mutations in the epidermal growth factor receptor (EGFR) are drivers for a subset of lung cancers. Osimertinib is a third-generation tyrosine kinase inhibitor (TKI) recently approved for the treatment of T790M-positive non-small cell lung cancer (NSCLC); however, acquired resistance to osimertinib is evident and resistance mechanisms remain incompletely defined. The EGFR G724S mutation was detected using hybrid-capture based comprehensive genomic profiling (CGP) and a hybrid-capture based circulating tumor DNA (ctDNA) assays in two cases of EGFR-driven lung adenocarcinoma in patients who had progressed on osimertinib treatment. This study demonstrates the importance of both tissue and blood based hybrid-capture based genomic profiling at disease progression to identifying novel resistance mechanisms in the clinic.

Collaboration


Dive into the V.A. Miller's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

J.S. Ross

Albany Medical College

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jeffrey S. Ross

State University of New York Upstate Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge