Network


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

Hotspot


Dive into the research topics where P.J. Stephens is active.

Publication


Featured researches published by P.J. Stephens.


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 Hematology & Oncology | 2014

Targeted therapy by combined inhibition of the RAF and mTOR kinases in malignant spindle cell neoplasm harboring the KIAA1549-BRAF fusion protein

Vivek Subbiah; Shannon N. Westin; Kai Wang; Dejka M. Araujo; Wei Lien Wang; Vincent A. Miller; Jeffrey S. Ross; P.J. Stephens; Gary A. Palmer; Siraj M. Ali

BackgroundOncologic patients who are extreme responders to molecularly targeted therapy provide an important opportunity to better understand the biologic basis of response and, in turn, inform clinical decision making. Malignant neoplasms with an uncertain histologic and immunohistochemical characterization present challenges both on initial diagnostic workups and then later in management, as current treatment algorithms are based on a morphologic diagnosis. Herein, we report a case of a difficult to characterize sarcoma-like lesion for which genomic profiling with clinical next generation sequencing (NGS) identified the molecular underpinnings of arrested progression(stable disease) under combination targeted therapy within a phase I clinical trial.MethodsGenomic profiling with clinical next generation sequencing was performed on the FoundationOne™ platform (Foundation Medicine, Cambridge MA). Histopathology and immunohistochemical studies were performed in the Department of Pathology, MD Anderson Cancer Center (Houston, TX). Treatment was administered in the context of a phase I clinical trial ClinicalTrials.gov Identifier: (NCT01187199).ResultsThe histology of the tumor was that of a spindle cell neoplasm, grade 2 by FNCLCC standards. Immunohistochemical staining was positive for S100 and CD34. Genomic profiling identified the following alterations: a KIAA1549-BRAF gene fusion resulting from a tandem duplication event, a homozygous deletion of PTEN, and frameshift insertion/deletions in CDKN2A A68fs*51, SUFU E283fs*3, and MAP3K1 N325fs*3. The patient had a 25% reduction in tumor (RECIST v1.1) following combination therapy consisting of sorafenib, temsirolimus, and bevazicumab within a phase I clinical trial.ConclusionsThe patient responded to combination targeted therapy that fortuitously targeted KIAA1549-BRAF and PTEN loss within a spindle cell neoplasm, as revealed by genomic profiling based on NGS. This is the first report of a tumor driven by a KIAA1549-BRAF fusion responding to sorafenib-based combination therapy.


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.


Science Translational Medicine | 2016

Triple-negative breast cancers with amplification of JAK2 at the 9p24 locus demonstrate JAK2-specific dependence

Justin M. Balko; Luis J. Schwarz; Na Luo; Monica V. Estrada; Jennifer M. Giltnane; Daniel Davila-Gonzalez; Kai Wang; Violeta Sanchez; Phillip T. Dean; Susan E. Combs; Donna Hicks; Joseph A. Pinto; Melissa D. Landis; Franco Doimi; Roman Yelensky; Vincent A. Miller; P.J. Stephens; David L. Rimm; Henry Gomez; Jenny C. Chang; Melinda E. Sanders; Rebecca S. Cook; Carlos L. Arteaga

JAK2 amplification decreases survival in triple-negative breast cancer but can be targeted with JAK2-specific inhibitors. Playing with JAKs Janus kinase (JAK) proteins are well known to be involved in cancer progression, and drugs such as ruxolitinib target these proteins, specifically JAK1 and JAK2. Balko et al. demonstrated frequent amplification of JAK2 in triple-negative breast cancer, a particularly aggressive and deadly form of the disease and showed that it was associated with decreased survival. The authors observed that JAK2 inhibition was effective in treating this type of breast cancer in mouse models. They also found that inhibiting JAK1 along with JAK2 in this context rendered the treatment ineffective, explaining why ruxolitinib does not work in triple-negative breast cancer and suggesting that specific JAK2 inhibitors may be a better approach. Amplifications at 9p24 have been identified in breast cancer and other malignancies, but the genes within this locus causally associated with oncogenicity or tumor progression remain unclear. Targeted next-generation sequencing of postchemotherapy triple-negative breast cancers (TNBCs) identified a group of 9p24-amplified tumors, which contained focal amplification of the Janus kinase 2 (JAK2) gene. These patients had markedly inferior recurrence-free and overall survival compared to patients with TNBC without JAK2 amplification. Detection of JAK2/9p24 amplifications was more common in chemotherapy-treated TNBCs than in untreated TNBCs or basal-like cancers, or in other breast cancer subtypes. Similar rates of JAK2 amplification were confirmed in patient-derived TNBC xenografts. In patients for whom longitudinal specimens were available, JAK2 amplification was selected for during neoadjuvant chemotherapy and eventual metastatic spread, suggesting a role in tumorigenicity and chemoresistance, phenotypes often attributed to a cancer stem cell–like cell population. In TNBC cell lines with JAK2 copy gains or amplification, specific inhibition of JAK2 signaling reduced mammosphere formation and cooperated with chemotherapy in reducing tumor growth in vivo. In these cells, inhibition of JAK1–signal transducer and activator of transcription 3 (STAT3) signaling had little effect or, in some cases, counteracted JAK2-specific inhibition. Collectively, these results suggest that JAK2-specific inhibitors are more efficacious than dual JAK1/2 inhibitors against JAK2-amplified TNBCs. Furthermore, JAK2 amplification is a potential biomarker for JAK2 dependence, which, in turn, can be used to select patients for clinical trials with JAK2 inhibitors.


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.


Science Translational Medicine | 2017

Genomic profiling of ER+ breast cancers after short-term estrogen suppression reveals alterations associated with endocrine resistance

Jennifer M. Giltnane; Katherine E. Hutchinson; Thomas Stricker; Luigi Formisano; Christian D. Young; Monica V. Estrada; Mellissa J. Nixon; Liping Du; Violeta Sanchez; Paula I. Gonzalez Ericsson; Maria G. Kuba; Melinda E. Sanders; Xinmeng J. Mu; Eliezer M. Van Allen; Nikhil Wagle; Ingrid A. Mayer; Vandana G. Abramson; Henry Gόmez; Monica Rizzo; Weiyi Toy; Sarat Chandarlapaty; Erica L. Mayer; Jason Christiansen; Danielle Murphy; Kerry Fitzgerald; Kai Wang; Jeffrey S. Ross; Vincent A. Miller; P.J. Stephens; Roman Yelensky

Genomic profiling of ER+/HER2− breast tumors after short-term estrogen deprivation revealed alterations associated with intrinsic resistance and provided mechanistic insights. A patient look at cancer resistance A variety of drugs that inhibit estrogen signaling are in use for breast cancer, but patients often develop resistance to these treatments. To understand how this resistance develops, Giltnane et al. evaluated 143 patients who were receiving the aromatase inhibitor letrozole to block estrogen signaling before undergoing surgery for breast cancer. By performing genomic analysis on these patients’ tumors, the authors were able to identify not only changes in gene expression and estrogen receptor gene fusions that correlated with resistance to therapy but also potential leads for future treatments that could help overcome this resistance. Inhibition of proliferation in estrogen receptor–positive (ER+) breast cancers after short-term antiestrogen therapy correlates with long-term patient outcome. We profiled 155 ER+/human epidermal growth factor receptor 2–negative (HER2−) early breast cancers from 143 patients treated with the aromatase inhibitor letrozole for 10 to 21 days before surgery. Twenty-one percent of tumors remained highly proliferative, suggesting that these tumors harbor alterations associated with intrinsic endocrine therapy resistance. Whole-exome sequencing revealed a correlation between 8p11-12 and 11q13 gene amplifications, including FGFR1 and CCND1, respectively, and high Ki67. We corroborated these findings in a separate cohort of serial pretreatment, postneoadjuvant chemotherapy, and recurrent ER+ tumors. Combined inhibition of FGFR1 and CDK4/6 reversed antiestrogen resistance in ER+ FGFR1/CCND1 coamplified CAMA1 breast cancer cells. RNA sequencing of letrozole-treated tumors revealed the existence of intrachromosomal ESR1 fusion transcripts and increased expression of gene signatures indicative of enhanced E2F-mediated transcription and cell cycle processes in cancers with high Ki67. These data suggest that short-term preoperative estrogen deprivation followed by genomic profiling can be used to identify druggable alterations that may cause intrinsic endocrine therapy resistance.


Blood Cancer Journal | 2017

Bi-allelic inactivation is more prevalent at relapse in multiple myeloma, identifying RB1 as an independent prognostic marker

Shweta S. Chavan; Jie He; Ruslana Tytarenko; Shayu Deshpande; Purvi Patel; Mark Bailey; Caleb K. Stein; Owen Stephens; Niels Weinhold; Nathan Petty; Douglas Steward; Leo Rasche; Michael Bauer; Cody Ashby; Erich Allen Peterson; Siraj M. Ali; Jeff Ross; Vincent A. Miller; P.J. Stephens; Sharmilan Thanendrarajan; Carolina Schinke; Maurizio Zangari; F van Rhee; B Barlogie; Tariq I. Mughal; Faith E. Davies; Gareth J. Morgan; Brian A. Walker

The purpose of this study is to identify prognostic markers and treatment targets using a clinically certified sequencing panel in multiple myeloma. We performed targeted sequencing of 578 individuals with plasma cell neoplasms using the FoundationOne Heme panel and identified clinically relevant abnormalities and novel prognostic markers. Mutational burden was associated with maf and proliferation gene expression groups, and a high-mutational burden was associated with a poor prognosis. We identified homozygous deletions that were present in multiple myeloma within key genes, including CDKN2C, RB1, TRAF3, BIRC3 and TP53, and that bi-allelic inactivation was significantly enriched at relapse. Alterations in CDKN2C, TP53, RB1 and the t(4;14) were associated with poor prognosis. Alterations in RB1 were predominantly homozygous deletions and were associated with relapse and a poor prognosis which was independent of other genetic markers, including t(4;14), after multivariate analysis. Bi-allelic inactivation of key tumor suppressor genes in myeloma was enriched at relapse, especially in RB1, CDKN2C and TP53 where they have prognostic significance.


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.

Collaboration


Dive into the P.J. Stephens's collaboration.

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
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge