Network


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

Hotspot


Dive into the research topics where Jonathan W. Riess is active.

Publication


Featured researches published by Jonathan W. Riess.


Nature Medicine | 2002

siRNA-directed inhibition of HIV-1 infection.

Carl D. Novina; Michael F. Murray; Derek M. Dykxhoorn; Paul J. Beresford; Jonathan W. Riess; Sang Kyung Lee; Ronald G. Collman; Judy Lieberman; Premlata Shankar; Phillip A. Sharp

RNA interference silences gene expression through short interfering 21–23-mer double-strand RNA segments that guide mRNA degradation in a sequence-specific fashion. Here we report that siRNAs inhibit virus production by targeting the mRNAs for either the HIV-1 cellular receptor CD4, the viral structural Gag protein or green fluorescence protein substituted for the Nef regulatory protein. siRNAs effectively inhibit pre- and/or post-integration infection events in the HIV-1 life cycle. Thus, siRNAs may have potential for therapeutic intervention in HIV-1 and other viral infections.


Cancer Discovery | 2013

A Drug Repositioning Approach Identifies Tricyclic Antidepressants as Inhibitors of Small Cell Lung Cancer and Other Neuroendocrine Tumors

Nadine S. Jahchan; Joel T. Dudley; Pawel K. Mazur; Natasha M. Flores; Dian Yang; Alec Palmerton; Anne Flore Zmoos; Dedeepya Vaka; Kim Q.t. Tran; Margaret Zhou; Karolina Krasinska; Jonathan W. Riess; Joel W. Neal; Purvesh Khatri; Kwon S. Park; Atul J. Butte; Julien Sage

UNLABELLED Small cell lung cancer (SCLC) is an aggressive neuroendocrine subtype of lung cancer with high mortality. We used a systematic drug repositioning bioinformatics approach querying a large compendium of gene expression profiles to identify candidate U.S. Food and Drug Administration (FDA)-approved drugs to treat SCLC. We found that tricyclic antidepressants and related molecules potently induce apoptosis in both chemonaïve and chemoresistant SCLC cells in culture, in mouse and human SCLC tumors transplanted into immunocompromised mice, and in endogenous tumors from a mouse model for human SCLC. The candidate drugs activate stress pathways and induce cell death in SCLC cells, at least in part by disrupting autocrine survival signals involving neurotransmitters and their G protein-coupled receptors. The candidate drugs inhibit the growth of other neuroendocrine tumors, including pancreatic neuroendocrine tumors and Merkel cell carcinoma. These experiments identify novel targeted strategies that can be rapidly evaluated in patients with neuroendocrine tumors through the repurposing of approved drugs. SIGNIFICANCE Our work shows the power of bioinformatics-based drug approaches to rapidly repurpose FDA-approved drugs and identifies a novel class of molecules to treat patients with SCLC, a cancer for which no effective novel systemic treatments have been identified in several decades. In addition, our experiments highlight the importance of novel autocrine mechanisms in promoting the growth of neuroendocrine tumor cells.


Nature Medicine | 2015

RAS-MAPK dependence underlies a rational polytherapy strategy in EML4-ALK–positive lung cancer

Gorjan Hrustanovic; Victor Olivas; Evangelos Pazarentzos; Asmin Tulpule; Saurabh Asthana; Collin M. Blakely; Ross A. Okimoto; Luping Lin; Dana S. Neel; Amit J. Sabnis; Jennifer Flanagan; Elton Chan; Marileila Varella-Garcia; Dara L. Aisner; Aria Vaishnavi; Sai-Hong Ignatius Ou; Eric A. Collisson; Eiki Ichihara; Philip C. Mack; Christine M. Lovly; Niki Karachaliou; Rafael Rosell; Jonathan W. Riess; Robert C. Doebele; Trever G. Bivona

One strategy for combating cancer-drug resistance is to deploy rational polytherapy up front that suppresses the survival and emergence of resistant tumor cells. Here we demonstrate in models of lung adenocarcinoma harboring the oncogenic fusion of ALK and EML4 that the GTPase RAS–mitogen-activated protein kinase (MAPK) pathway, but not other known ALK effectors, is required for tumor-cell survival. EML4-ALK activated RAS-MAPK signaling by engaging all three major RAS isoforms through the HELP domain of EML4. Reactivation of the MAPK pathway via either a gain in the number of copies of the gene encoding wild-type K-RAS (KRASWT) or decreased expression of the MAPK phosphatase DUSP6 promoted resistance to ALK inhibitors in vitro, and each was associated with resistance to ALK inhibitors in individuals with EML4-ALK–positive lung adenocarcinoma. Upfront inhibition of both ALK and the kinase MEK enhanced both the magnitude and duration of the initial response in preclinical models of EML4-ALK lung adenocarcinoma. Our findings identify RAS-MAPK dependence as a hallmark of EML4-ALK lung adenocarcinoma and provide a rationale for the upfront inhibition of both ALK and MEK to forestall resistance and improve patient outcomes.


Lung Cancer | 2012

ERCC1 expression in circulating tumor cells (CTCs) using a novel detection platform correlates with progression-free survival (PFS) in patients with metastatic non-small-cell lung cancer (NSCLC) receiving platinum chemotherapy.

Millie Das; Jonathan W. Riess; Paul Frankel; Erich J. Schwartz; Robyn Bennis; H. Ben Hsieh; Xiaohe Liu; Janey C. Ly; Lisa Zhou; Jorge Nieva; Heather A. Wakelee; Richard H. Bruce

PURPOSE To utilize a novel circulating tumor cell (CTC) technology to quantify ERCC1 expression on CTCs and determine whether ERCC1 expression levels predict efficacy of platinum-based chemotherapy in patients with metastatic non-small-cell lung cancer (NSCLC). EXPERIMENTAL DESIGN ERCC1 expression was measured in 17 metastatic NSCLC patients who received platinum-based therapy and had ≥2 intact CTCs with acceptable ERCC1 expression assay results. ERCC1 levels were determined from average expression on individual CTCs in each sample. Progression-free survival (PFS) was calculated from the date of therapy initiation. RESULTS PFS decreased with increasing ERCC1 expression (p<0.04, F-test, linear regression). Lack of ERCC1 expression was associated with longer PFS (266 days versus 172 days, log-rank, p<0.02) in a Kaplan-Meier analysis using ERCC expression level of 1 as a cutoff (range 0-30). The difference in survival was statistically significant with a hazard ratio of 4.20 (95% CI 1.25-14.1, p<0.02, log-rank). PFS was also observed to decrease with increased cytokeratin (CK) expression (p<0.01 long-rank (Cox regression) and F-test (linear regression)). The hazard ratio is 4.38 (95% CI 1.76-10.9) for each log-change in CK value until progression was noted on imaging. CONCLUSION Low expression of ERCC1 on CTCs correlates with PFS in patients with metastatic NSCLC receiving platinum-based therapy.


Journal of Thoracic Oncology | 2015

Diffuse High Intensity PD–L1 Staining in Thymic Epithelial Tumors

Sukhmani K. Padda; Jonathan W. Riess; Erich J. Schwartz; Lu Tian; Holbrook Kohrt; Joel W. Neal; Robert B. West; Heather A. Wakelee

Introduction: Blockade of the immune checkpoint programmed death receptor ligand-1 (PD-L1)/PD-1 pathway has well-established clinical activity across many tumor types. PD-L1 protein expression by immunohistochemistry is emerging as a predictive biomarker of response to these therapies. Here, we examine PD-L1 expression in a thymic epithelial tumor (TET) tissue microarray (TMA). Methods: The TMA contained 69 TETs and 17 thymic controls, with each case represented by triplicate cores. The TMA was stained with rabbit monoclonal antibody (clone 15; Sino Biological, Beijing, China) to human PD-L1. PD-L1 staining was scored based on intensity as follows: 0 = none, 1 = equivocal/uninterpretable, 2 = weak, and 3 = intermediate-strong. Those cases with all cores scoring three in the epithelial component were categorized as PD-L1high and the remaining as PD-L1low. Results: PD-L1high scores were more frequent in TETs than in controls (68.1% versus 17.6%; p = 0.0036). PD-L1 scores and histology were significantly correlated, with higher intensity staining in World Health Organization (WHO). B2/B3/C TETs. Only 14.8% of TETs had PD-L1 staining of associated lymphocytes. In an adjusted analysis (age/sex), PD-L1high TETs had a significantly worse overall survival (hazard ratio: 5.40, 95% confidence interval: 1.13–25.89; p = 0.035) and a trend for worse event-free survival (hazard ratio: 2.94, 95% confidence interval: 0.94–9.24; p = 0.064). Conclusions: PD-L1 expression was present in all cases of TETs within the epithelial component but only in a minority in the lymphocytic component. TETs stained more intensely for PD-L1 than in controls, and PD-L1high TETs were associated with more aggressive histology and worse prognosis. This study lends rationale to a clinical trial with anti-PD-1/PD-L1 therapy in this rare tumor type.


Journal of Clinical Oncology | 2017

Targeting RET in Patients With RET-Rearranged Lung Cancers: Results From the Global, Multicenter RET Registry

Oliver Gautschi; Julie Milia; Thomas Filleron; Juergen Wolf; David P. Carbone; Dwight Owen; Ross Camidge; Vignhesh Narayanan; Robert C. Doebele; Benjamin Besse; Jordi Remon-Masip; Pasi A. Jänne; Mark M. Awad; Nir Peled; Chul Cho Byoung; Daniel D. Karp; Michael Van Den Heuvel; Heather A. Wakelee; Joel W. Neal; Tony Mok; James Chih-Hsin Yang; Sai-Hong Ignatius Ou; Georg Pall; Patrizia Froesch; G. Zalcman; David R. Gandara; Jonathan W. Riess; Vamsidhar Velcheti; Kristin Zeidler; Joachim Diebold

Purpose In addition to prospective trials for non-small-cell lung cancers (NSCLCs) that are driven by less common genomic alterations, registries provide complementary information on patient response to targeted therapies. Here, we present the results of an international registry of patients with RET-rearranged NSCLCs, providing the largest data set, to our knowledge, on outcomes of RET-directed therapy thus far. Methods A global, multicenter network of thoracic oncologists identified patients with pathologically confirmed NSCLC that harbored a RET rearrangement. Molecular profiling was performed locally by reverse transcriptase polymerase chain reaction, fluorescence in situ hybridization, or next-generation sequencing. Anonymized data-clinical, pathologic, and molecular features-were collected centrally and analyzed by an independent statistician. Best response to RET tyrosine kinase inhibition administered outside of a clinical trial was determined by RECIST v1.1. Results By April 2016, 165 patients with RET-rearranged NSCLC from 29 centers across Europe, Asia, and the United States were accrued. Median age was 61 years (range, 29 to 89 years). The majority of patients were never smokers (63%) with lung adenocarcinomas (98%) and advanced disease (91%). The most frequent rearrangement was KIF5B-RET (72%). Of those patients, 53 received one or more RET tyrosine kinase inhibitors in sequence: cabozantinib (21 patients), vandetanib (11 patients), sunitinib (10 patients), sorafenib (two patients), alectinib (two patients), lenvatinib (two patients), nintedanib (two patients), ponatinib (two patients), and regorafenib (one patient). The rate of any complete or partial response to cabozantinib, vandetanib, and sunitinib was 37%, 18%, and 22%, respectively. Further responses were observed with lenvantinib and nintedanib. Median progression-free survival was 2.3 months (95% CI, 1.6 to 5.0 months), and median overall survival was 6.8 months (95% CI, 3.9 to 14.3 months). Conclusion Available multikinase inhibitors had limited activity in patients with RET-rearranged NSCLC in this retrospective study. Further investigation of the biology of RET-rearranged lung cancers and identification of new targeted therapeutics will be required to improve outcomes for these patients.


Clinical Lung Cancer | 2014

Acquired resistance to targeted therapies against oncogene-driven non-small-cell lung cancer: Approach to subtyping progressive disease and clinical implications

David R. Gandara; Tianhong Li; Primo N. Lara; Karen Kelly; Jonathan W. Riess; Mary W. Redman; Philip C. Mack

In the emerging era of targeted therapy for advanced-stage non-small-cell lung cancer, it is becoming increasingly important to anticipate underlying driver oncogene alterations at the time of initial diagnosis and tumor-tissue acquisition, so that patients can be selected in a timely fashion for first-line tyrosine kinase inhibitor (TKI) therapy if their cancers are found to harbor tyrosine-kinase-activating mutations in the epidermal growth factor receptor gene or gain-of-function rearrangements in the anaplastic lymphoma kinase gene. However, despite the clear benefits of TKI therapy over chemotherapy in these settings, the eventual emergence of acquired resistance and progressive disease (PD) is universal. How to best approach oncogene-driven non-small-cell lung cancer at the time of acquired resistance to initial TKI therapy is an increasingly complex question because of variability in mechanisms of resistance, extent of PD, and inter- and intrapatient tumor heterogeneity. Here we propose an approach to subtyping PD in the setting of acquired resistance as well as subsequent clinical implications.


Clinical Lung Cancer | 2014

Prolonged survival of patients with non-small-cell lung cancer with leptomeningeal carcinomatosis in the modern treatment era.

Jonathan W. Riess; Seema Nagpal; Michael; Michael Zeineh; Matthew A. Gubens; Kavitha Ramchandran; Joel W. Neal; Heather A. Wakelee

INTRODUCTION Leptomeningeal carcinomatosis (LM) is a severe complication of non-small-cell lung cancer (NSCLC) historically associated with poor prognosis. New chemotherapeutic and targeted treatments could potentially affect the natural history of LM. PATIENTS AND METHODS Patients with a pathologic diagnosis of NSCLC with LM treated at Stanford between 2003 and 2011 were identified via institutional databases and medical records. LM was defined by cerebrospinal fluid (CSF) that was positive for malignant cells or by LM enhancement on magnetic resonance imaging with gadolinium contrast. Retrospective, landmark analyses were performed to estimate survival. Statistical analyses were performed using SAS Enterprise Guide, version 4.3. RESULTS LM was identified in 30 patients. All cases were adenocarcinoma; 60% of patients had a known or suspected driver mutation. The mean age was 58 years. Of the 30 patients, 67% were women; 70% were nonsmokers; 27% initially presented with LM; 84% received systemic treatment at or after development of LM; and 53% of these patients received modern systemic therapy for their LM, defined as a regimen containing pemetrexed, bevacizumab, or a tyrosine kinase inhibitor. Mean overall survival after LM diagnosis was 6 months (95% CI, 3-12). Patients who received modern systemic therapy for LM had decreased hazard of death (hazard ratio [HR], 0.24; P = .007). CONCLUSION In this retrospective, single-institution analysis, median survival with LM was higher compared with historical experience. Patients who received modern systemic therapy for their LM had particularly good outcomes. These data provide evidence for improving survival outcomes in the modern treatment era for this difficult-to-treat complication.


Clinical Lung Cancer | 2013

A Case Series of Lengthy Progression-Free Survival With Pemetrexed-Containing Therapy in Metastatic Non–Small-Cell Lung Cancer Patients Harboring ROS1 Gene Rearrangements

Jonathan W. Riess; Sukhmani K. Padda; Charles D. Bangs; Millie Das; Joel W. Neal; Adour R. Adrouny; Athena M. Cherry; Heather A. Wakelee

c-ros oncogene 1 (ROS1), an orphan receptor tyrosine kinase, is a recently defined molecular subset of lung cancer identified in approximately 1% to 2% of screened patients.1,2 Like anaplastic lymphoma receptor tyrosine kinase (ALK) translocations, ROS1 rearrangements lead to constitutive kinase activation and commonly arise in young nonsmokers with adenocarcinoma tumor histology.3 Crizotinib (Pfizer), recently approved by the US Food and Drug Administration for treatment of metastatic non–small-cell lung cancer (NSCLC) patients with ALK rearranged tumors, has potent activity in cell lines with ROS1 translocations and recently demonstrated clinical activity in patients whose tumors harbor ROS1 rearrangements.3,4 Because of the recently discovered importance of ROS1 trans-locations in a small subset of NSCLC, we know little regarding the response to specific chemotherapeutics in patients whose tumors harbor these molecular alterations. Pemetrexed (Lilly) inhibits thymidylate synthase (TS) and other folate-dependent enzymes, and is a chemotherapeutic agent commonly used to treat nonsquamous NSCLC.5 Our institution conducted a retrospective case series of all meta-static lung cancer patients with ROS1 rearrangements. We recently began testing for ROS1 gene rearrangements using break-apart fluorescence in situ hybridization (FISH) in patients with lung adenocarcinoma and no other known mutations. A total of 129 ROS1 assays using FISH have been completed at Stanford with 5 ROS1 rearrangements identified; 1 additional patient treated at Stanford tested positive using FISH at an outside institution. Four of the 6 patients with detectable ROS1 rearrangements have metastatic cancer; and all 4 patients have a strikingly long progression-free survival (PFS) using pemetrexed—given as either single-agent second-line treatment or as part of front-line chemotherapy (Table 1). Table 1 Summary of ROS1 Cases Case 1 The patient is a 35-year-old never smoking woman who presented with dyspnea. Imaging revealed confluent pulmonary nodules in the left lung with a fluorine-18 fluorodeoxyglucose-avid pleural effusion. Mediastinoscopy showed N3 disease with right-sided mediastinal nodes positive for lung adenocarcinoma. The patient received 6 cycles of carboplatin, pemetrexed, and bevacizumab with a partial response followed by continuation maintenance with pemetrexed and bevacizumab for 21 cycles (Figure 1). During maintenance treatment, FISH was performed and revealed a ROS1 rearrangement. Bevacizumab was eventually discontinued because of nonspecific neurologic complaints. She continues on pemetrexed alone as maintenance therapy for > 14 cycles with minimal disease burden. Total time using pemetrexed is now > 36 months. Figure 1 Computed Tomography Scan of a NSCLC Patient With a ROS1 Gene Rearrangement Before Treatment (Left) and More Than 26 Months Later (Right) During Pemetrexed Continuation Maintenance Case 2 The patient is a 64-year-old man with a distant light smoking history who presented with a palpable left supraclavicular lymph node. Biopsy of this node revealed lung adenocarcinoma. The patients NSCLC was initially stage IIIB and he was treated with concurrent chemoradiation with carboplatin and paclitaxel. He developed biopsy-proven recurrence of disease 1 year later with multiple bilateral pulmonary nodules. He then received 6 cycles of carboplatin, pemetrexed, and bevacizumab with a partial response, followed by continuation maintenance pemetrexed and bevacizumab for 37 cycles. During maintenance treatment, a ROS1 rearrangement was detected by FISH. Proteinuria eventually led to discontinuation of bevacizumab. He continues taking pemetrexed alone as maintenance therapy for additional > 19 cycles. Total time taking pemetrexed is now > 47 months.


British Journal of Cancer | 2014

A phase II multicentre study of ziv-aflibercept in combination with cisplatin and pemetrexed in patients with previously untreated advanced/metastatic non-squamous non-small cell lung cancer

Hongbin Chen; Manuel R. Modiano; Joel W. Neal; Julie R. Brahmer; James R. Rigas; Robert M. Jotte; N. Leighl; Jonathan W. Riess; Calvin J. Kuo; L Liu; B Gao; A T DiCioccio; Alex A. Adjei; Heather A. Wakelee

Background:This study evaluated the efficacy and safety of ziv-aflibercept in combination with cisplatin and pemetrexed in non-small cell lung cancer (NSCLC).Methods:This single arm, multicentre phase II trial enrolled patients with previously untreated, locally advanced or metastatic non-squamous NSCLC. Patients received intravenous ziv-aflibercept 6 mg kg−1, pemetrexed 500 mg m−2, and cisplatin 75 mg m−2, every 21 days for up to six cycles. Maintenance administration of ziv-aflibercept was to continue until disease progression, intolerable toxicity or other cause for withdrawal. The co-primary end points were objective response rate (ORR) and progression-free survival (PFS). Planned sample size was 72 patients.Results:The study was closed prematurely because of three confirmed and two suspected cases of reversible posterior leukoencephalopathy syndrome (RPLS). A total of 42 patients were enrolled. Median age was 61.5 years; 55% were male, 86% Caucasian and 50% had Eastern Cooperative Oncology Group performance status (ECOG PS)=0. A median of four cycles of ziv-aflibercept was administered. The most common treatment-emergent adverse events (TEAEs) of any grade were nausea (69%) and fatigue (67%), with hypertension (36%) as the most common grade 3/4 TEAE. Of the 38 evaluable patients, ORR was 26% and median PFS was 5 months.Conclusion:Cases of RPLS had been observed in other studies in the ziv-aflibercept clinical development programme but the rate observed in this study was higher than previously observed. This might be related to declining renal function and/or hypertension. Although ORR and PFS were in accordance with most historical first-line NSCLC studies, this combination of ziv-aflibercept/cisplatin/pemetrexed will not be further explored in NSCLC.

Collaboration


Dive into the Jonathan W. Riess's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Philip C. Mack

University of California

View shared research outputs
Top Co-Authors

Avatar

Primo N. Lara

University of California

View shared research outputs
Top Co-Authors

Avatar

Karen Kelly

Anschutz Medical Campus

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robert C. Doebele

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Tianhong Li

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge