276 results on '"Fanta, Paul T"'
Search Results
2. Establishment of Patient-derived Succinate Dehydrogenase-deficient Gastrointestinal Stromal Tumor Models For Predicting Therapeutic Response
- Author
-
Yebra, Mayra, Bhargava, Shruti, Kumar, Avi, Burgoyne, Adam M, Tang, Chih-Min, Yoon, Hyunho, Banerjee, Sudeep, Aguilera, Joseph, Cordes, Thekla, Sheth, Vipul, Noh, Sangkyu, Ustoy, Rowan, Li, Sam, Advani, Sunil J, Corless, Christopher L, Heinrich, Michael C, Kurzrock, Razelle, Lippman, Scott M, Fanta, Paul T, Harismendy, Olivier, Metallo, Christian, and Sicklick, Jason K
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Oncology and Carcinogenesis ,Rare Diseases ,Digestive Diseases ,Pediatric ,Genetics ,Cancer ,Clinical Research ,Orphan Drug ,2.1 Biological and endogenous factors ,Good Health and Well Being ,Adolescent ,Gastrointestinal Neoplasms ,Gastrointestinal Stromal Tumors ,Humans ,Mutation ,Proto-Oncogene Proteins c-kit ,Receptor ,Platelet-Derived Growth Factor alpha ,Succinate Dehydrogenase ,Young Adult ,Oncology & Carcinogenesis ,Clinical sciences ,Oncology and carcinogenesis - Abstract
PurposeGastrointestinal stromal tumor (GIST) is the most common sarcoma of the gastrointestinal tract, with mutant succinate dehydrogenase (SDH) subunits (A-D) comprising less than 7.5% (i.e., 150-200/year) of new cases annually in the United States. Contrary to GISTs harboring KIT or PDGFRA mutations, SDH-mutant GISTs affect adolescents/young adults, often metastasize, and are frequently resistant to tyrosine kinase inhibitors (TKI). Lack of human models for any SDH-mutant tumors, including GIST, has limited molecular characterization and drug discovery.Experimental designWe describe methods for establishing novel patient-derived SDH-mutant (mSDH) GIST models and interrogated the efficacy of temozolomide on these tumor models in vitro and in clinical trials of patients with mSDH GIST.ResultsMolecular and metabolic characterization of our patient-derived mSDH GIST models revealed that these models recapitulate the transcriptional and metabolic hallmarks of parent tumors and SDH deficiency. We further demonstrate that temozolomide elicits DNA damage and apoptosis in our mSDH GIST models. Translating our in vitro discovery to the clinic, a cohort of patients with SDH-mutant GIST treated with temozolomide (n = 5) demonstrated a 40% objective response rate and 100% disease control rate, suggesting that temozolomide represents a promising therapy for this subset of GIST.ConclusionsWe report the first methods to establish patient-derived mSDH tumor models, which can be readily employed for understanding patient-specific tumor biology and treatment strategies. We also demonstrate that temozolomide is effective in patients with mSDH GIST who are refractory to existing chemotherapeutic drugs (namely, TKIs) in clinic for GISTs, bringing a promising treatment option for these patients to clinic.See related commentary by Blakely et al., p. 3.
- Published
- 2022
3. Comprehensive genomic landscape and precision therapeutic approach in biliary tract cancers
- Author
-
Okamura, Ryosuke, Kurzrock, Razelle, Mallory, Robert J, Fanta, Paul T, Burgoyne, Adam M, Clary, Bryan M, Kato, Shumei, and Sicklick, Jason K
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Oncology and Carcinogenesis ,Rare Diseases ,Clinical Research ,Biotechnology ,Human Genome ,Genetics ,Cancer ,Digestive Diseases ,Cancer Genomics ,Precision Medicine ,5.1 Pharmaceuticals ,Good Health and Well Being ,Adult ,Aged ,Aged ,80 and over ,Antineoplastic Agents ,Biliary Tract Neoplasms ,Circulating Tumor DNA ,Class I Phosphatidylinositol 3-Kinases ,Cyclin-Dependent Kinase Inhibitor p15 ,Cyclin-Dependent Kinase Inhibitor p16 ,DNA ,Neoplasm ,Feasibility Studies ,Female ,High-Throughput Nucleotide Sequencing ,Humans ,Male ,Middle Aged ,Proto-Oncogene Proteins p21(ras) ,Sequence Analysis ,DNA ,Survival Analysis ,Treatment Outcome ,Tumor Suppressor Protein p53 ,biliary tract cancers ,biomarker ,cholangiocarcinoma ,circulating tumor DNA ,liquid biopsy ,molecular profiling ,personalized cancer therapy ,Oncology & Carcinogenesis ,Oncology and carcinogenesis - Abstract
Biliary tract cancers have dismal prognoses even when cytotoxic chemotherapy is administered. There is an unmet need to develop precision treatment approaches using comprehensive genomic profiling. A total of 121 patients with biliary tract cancers were analyzed for circulating-tumor DNA (ctDNA) and/or tissue-based tumor DNA (tissue-DNA) using clinical-grade next-generation sequencing: 71 patients (59%) had ctDNA; 90 (74%), tissue-DNA; and 40 (33%), both. Efficacy of targeted therapeutic approaches was assessed based upon ctDNA and tissue-DNA. At least one characterized alteration was detected in 76% of patients (54/71) for ctDNA [median, 2 (range, 0-9)] and 100% (90/90) for tissue-DNA [median, 4 (range, 1-9)]. Most common alterations occurred in TP53 (38%), KRAS (28%), and PIK3CA (14%) for ctDNA vs TP53 (44%), CDKN2A/B (33%) and KRAS (29%) for tissue-DNA. In 40 patients who had both ctDNA and tissue-DNA sequencing, overall concordance was higher between ctDNA and metastatic site tissue-DNA than between ctDNA and primary tumor DNA (78% vs 65% for TP53, 100% vs 74% for KRAS and 100% vs 87% for PIK3CA [But not statistical significance]). Among 80 patients who received systemic treatment, the molecularly matched therapeutic regimens based on genomic profiling showed a significantly longer progression-free survival (hazard ratio [95%confidence interval], 0.60 [0.37-0.99]. P = .047 [multivariate]) and higher disease control rate (61% vs 35%, P = .04) than unmatched regimens. Evaluation of ctDNA and tissue-DNA is feasible in biliary tract cancers.
- Published
- 2021
4. Real-world data from a molecular tumor board demonstrates improved outcomes with a precision N-of-One strategy.
- Author
-
Kato, Shumei, Kim, Ki Hwan, Lim, Hyo Jeong, Boichard, Amelie, Nikanjam, Mina, Weihe, Elizabeth, Kuo, Dennis J, Eskander, Ramez N, Goodman, Aaron, Galanina, Natalie, Fanta, Paul T, Schwab, Richard B, Shatsky, Rebecca, Plaxe, Steven C, Sharabi, Andrew, Stites, Edward, Adashek, Jacob J, Okamura, Ryosuke, Lee, Suzanna, Lippman, Scott M, Sicklick, Jason K, and Kurzrock, Razelle
- Subjects
Humans ,Neoplasms ,Antineoplastic Agents ,Disease-Free Survival ,Treatment Outcome ,Genome ,Human ,Adolescent ,Adult ,Aged ,Aged ,80 and over ,Middle Aged ,Child ,Child ,Preschool ,Female ,Male ,Young Adult ,Precision Medicine ,Circulating Tumor DNA ,Genome ,Human ,and over ,Preschool - Abstract
Next-generation sequencing (NGS) can identify novel cancer targets. However, interpreting the molecular findings and accessing drugs/clinical trials is challenging. Furthermore, many tumors show resistance to monotherapies. To implement a precision strategy, we initiated a multidisciplinary (basic/translational/clinical investigators, bioinformaticians, geneticists, and physicians from multiple specialties) molecular tumor board (MTB), which included a project manager to facilitate obtaining clinical-grade biomarkers (blood/tissue NGS, specific immunohistochemistry/RNA expression including for immune-biomarkers, per physician discretion) and medication-acquisition specialists/clinical trial coordinators/navigators to assist with medication access. The MTB comprehensively reviewed patient characteristics to develop N-of-One treatments implemented by the treating physician's direction under the auspices of a master protocol. Overall, 265/429 therapy-evaluable patients (62%) were matched to ≥1 recommended drug. Eighty-six patients (20%) matched to all drugs recommended by MTB, including combinatorial approaches, while 38% received physician's choice regimen, generally with unmatched approach/low degree of matching. Our results show that patients who receive MTB-recommended regimens (versus physician choice) have significantly longer progression-free (PFS) and overall survival (OS), and are better matched to therapy. High (≥50%) versus low (
- Published
- 2020
5. Prognostic implications of RAS alterations in diverse malignancies and impact of targeted therapies
- Author
-
Kato, Shumei, Okamura, Ryosuke, Sicklick, Jason K, Daniels, Gregory A, Hong, David S, Goodman, Aaron, Weihe, Elizabeth, Lee, Suzanna, Khalid, Noor, Collier, Rachel, Mareboina, Manvita, Riviere, Paul, Whitchurch, Theresa J, Fanta, Paul T, Lippman, Scott M, and Kurzrock, Razelle
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Precision Medicine ,Rare Diseases ,Genetics ,Cancer ,Health Disparities ,2.1 Biological and endogenous factors ,Aged ,Aged ,80 and over ,Antineoplastic Combined Chemotherapy Protocols ,California ,Female ,Humans ,MAP Kinase Signaling System ,Male ,Middle Aged ,Mitogen-Activated Protein Kinase Kinases ,Molecular Targeted Therapy ,Mutation ,Neoplasms ,Observational Studies as Topic ,Prognosis ,Progression-Free Survival ,Protein Kinase Inhibitors ,ras Proteins ,RAS ,next-generation sequencing ,targeted therapy ,Oncology & Carcinogenesis ,Oncology and carcinogenesis - Abstract
RAS alterations are often found in difficult-to-treat malignancies and are considered "undruggable." To better understand the clinical correlates and coaltered genes of RAS alterations, we used targeted next-generation sequencing (NGS) to analyze 1,937 patients with diverse cancers. Overall, 20.9% of cancers (405/1,937) harbored RAS alterations. Most RAS-altered cases had genomic coalterations (95.3%, median: 3, range: 0-51), often involving genes implicated in oncogenic signals: PI3K pathway (31.4% of 405 cases), cell cycle (31.1%), tyrosine kinase families (21.5%) and MAPK signaling (18.3%). Patients with RAS-altered versus wild-type RAS malignancies had significantly worse overall survival (OS; p = 0.02 [multivariate]), with KRAS alterations, in particular, showing shorter survival. Moreover, coalterations in both RAS and PI3K signaling or cell-cycle-associated genes correlated with worse OS (p = 0.004 and p
- Published
- 2020
6. Comprehensive Genomic Landscape and Precision Therapeutic Approach in Biliary Tract Cancers
- Author
-
Okamura, Ryosuke, Kurzrock, Razelle, Mallory, Robert J, Fanta, Paul T, Burgoyne, Adam M, Clary, Bryan M, Kato, Shumei, and Sicklick, Jason K
- Subjects
Digestive Diseases ,Cancer ,Genetics ,Human Genome ,Biotechnology ,Good Health and Well Being ,biliary tract cancer ,cholangiocarcinoma ,circulating tumor DNA ,liquid biopsy ,molecular oncology ,biomarker ,personalized cancer therapy - Published
- 2020
7. Revisiting Epidermal Growth Factor Receptor (EGFR) Amplification as a Target for Anti-EGFR Therapy: Analysis of Cell-Free Circulating Tumor DNA in Patients With Advanced Malignancies
- Author
-
Kato, Shumei, Okamura, Ryosuke, Mareboina, Manvita, Lee, Suzanna, Goodman, Aaron, Patel, Sandip P, Fanta, Paul T, Schwab, Richard B, Vu, Peter, Raymond, Victoria M, Lanman, Richard B, Sicklick, Jason K, Lippman, Scott M, and Kurzrock, Razelle
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Oncology and Carcinogenesis ,Biotechnology ,Human Genome ,Lung ,Cancer Genomics ,Cancer ,Genetics ,5.1 Pharmaceuticals ,Good Health and Well Being ,Clinical Research ,4.1 Discovery and preclinical testing of markers and technologies ,Oncology and carcinogenesis - Abstract
PurposeTo date, evidence for tissue epidermal growth factor receptor (EGFR) overexpression as a biomarker for anti-EGFR therapies has been weak. We investigated the genomic landscape of EGFR amplification in blood-derived cell-free tumor DNA (cfDNA) across diverse cancers and the role of anti-EGFR therapies in achieving response.MethodsWe assessed EGFR amplification status among 28,584 patients with malignancies evaluated by clinical-grade next-generation sequencing (NGS) of blood-derived cfDNA (54- to 73-gene panel). Furthermore, we curated the clinical characteristics of 1,434 patients at the University of California San Diego who had cfDNA testing by this NGS test.ResultsOverall, EGFR amplification was detected in cfDNA from 8.5% of patients (2,423 of 28,584), most commonly in colorectal (16.3% [458 of 2,807]), non-small-cell lung (9.0% [1,096 of 12,197]), and genitourinary cancers (8.1% [170 of 2,104]). Most patients had genomic coalterations (96.9% [95 of 98]), frequently involving genes affecting other tyrosine kinases (72.4% [71 of 98]), mitogen-activated protein kinase cascades (56.1% [55 of 98]), cell-cycle-associated signals (52.0% [51 of 98]), and the phosphoinositide 3-kinase pathway (35.7% [35 of 98]). EGFR amplification emerged in serial cfDNA after various anticancer therapies (n = 6), including checkpoint inhibitors (n = 4), suggesting a possible role for these amplifications in acquired resistance. Nine evaluable patients with EGFR amplification were treated with anti-EGFR-based regimens; five (55.6%) achieved partial responses, including three patients whose tissue NGS lacked EGFR amplification.ConclusionEGFR amplification was detected in cfDNA among 8.5% of 28,584 diverse cancers. Most patients had coexisting alterations. Responses were observed in five of nine patients who received EGFR inhibitors. Incorporating EGFR inhibitors into the treatment regimens of patients harboring EGFR amplification in cfDNA merits additional study.
- Published
- 2019
8. Genomic Assessment of Blood-Derived Circulating Tumor DNA in Patients With Colorectal Cancers: Correlation With Tissue Sequencing, Therapeutic Response, and Survival
- Author
-
Kato, Shumei, Schwaederlé, Maria C, Fanta, Paul T, Okamura, Ryosuke, Leichman, Lawrence, Lippman, Scott M, Lanman, Richard B, Raymond, Victoria M, Talasaz, AmirAli, and Kurzrock, Razelle
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Oncology and Carcinogenesis ,Precision Medicine ,Minority Health ,Cancer Genomics ,Genetics ,Cancer ,Biotechnology ,Human Genome ,Digestive Diseases ,Health Disparities ,Colo-Rectal Cancer ,Good Health and Well Being ,Oncology and carcinogenesis - Abstract
PurposeGenomic alterations in blood-derived circulating tumor DNA (ctDNA) from patients with colorectal cancers were correlated with clinical outcomes.Patients and methodsNext-generation sequencing of ctDNA (54- to 73-gene panel) was performed in 94 patients with colorectal cancer.ResultsMost patients (96%) had metastatic or recurrent disease at the time of blood draw. The median number of nonsynonymous alterations per patient was three (range, zero to 30). The most frequently aberrant genes were TP53 (52.1% of patients), KRAS (34%), and APC (28.7%). Concordance between tissue and blood next-generation sequencing ranged from 63.2% (APC) to 85.5% (BRAF). Altogether, 74 patients (79%) had one or more nonsynonymous alterations, 69 (73%) had one or more potentially actionable alterations, and 61 (65%) had an alteration actionable by a drug approved by the US Food and Drug Administration (on or off label). Lung metastases correlated with improved survival from diagnosis in univariable analysis. ctDNA of 5% or more from blood tests as well as EGFR and ERBB2 (HER2) nonsynonymous alterations correlated with worse survival (but only ERBB2 remained significant in multivariable analysis). No two patients had identical molecular portfolios. Overall, 65% versus 31% of patients treated with matched (n = 17) versus unmatched therapy (n = 18) after ctDNA testing achieved stable disease for 6 months or more, partial response, or complete response (P = .045); progression-free survival, 6.1 versus 2.3 months (P = .08); and survival not reached versus 9.4 months (P = .146; all by multivariable analysis).ConclusionPatients with colorectal cancer have heterogeneous ctDNA profiles, and most harbor potentially actionable ctDNA alterations. Matched therapy yielded higher rates of stable disease for 6 months or more, partial response, or complete response. ctDNA assessment may have clinical utility and merits further investigation.
- Published
- 2019
9. Genomic Profiling of Blood-Derived Circulating Tumor DNA from Patients with Colorectal Cancer: Implications for Response and Resistance to Targeted Therapeutics
- Author
-
Choi, In Sil, Kato, Shumei, Fanta, Paul T, Leichman, Lawrence, Okamura, Ryosuke, Raymond, Victoria M, Lanman, Richard B, Lippman, Scott M, and Kurzrock, Razelle
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Oncology and Carcinogenesis ,Digestive Diseases ,Biotechnology ,Colo-Rectal Cancer ,Cancer Genomics ,Genetics ,Cancer ,Human Genome ,Genetic Testing ,5.1 Pharmaceuticals ,Good Health and Well Being ,Adult ,Aged ,Aged ,80 and over ,Biomarkers ,Tumor ,Circulating Tumor DNA ,Colorectal Neoplasms ,Drug Resistance ,Neoplasm ,Female ,Genomics ,Humans ,Male ,Middle Aged ,Molecular Targeted Therapy ,Proto-Oncogene Proteins p21(ras) ,Pharmacology and Pharmaceutical Sciences ,Oncology & Carcinogenesis ,Biochemistry and cell biology ,Oncology and carcinogenesis - Abstract
Molecular profiling of circulating tumor DNA (ctDNA) is a promising noninvasive tool. Here, next-generation sequencing (NGS) of blood-derived ctDNA was performed in patients with advanced colorectal cancer. We investigated ctDNA-derived genomic alterations, including potential actionability, concordance with tissue NGS, and serial dynamics in 78 patients with colorectal cancer using a clinical-grade NGS assay that detects single nucleotide variants (54-73 genes) and selected copy-number variants, fusions, and indels. Overall, 63 patients [80.8% (63/78)] harbored ctDNA alterations; 59 [75.6% (59/78)], ≥1 characterized alteration (variants of unknown significance excluded). All 59 patients had actionable alterations potentially targetable with FDA-approved drugs [on-label and/or off-label (N = 54) or with experimental drugs in clinical trials (additional five patients); University of California San Diego Molecular Tumor Board assessment]: 45, by OncoKB (http://oncokb.org/#/). The tissue and blood concordance rates for common specific alterations ranged from 62.3% to 86.9% (median = 5 months between tests). In serial samples from patients on anti-EGFR therapy, multiple emerging alterations in genes known to be involved in therapeutic resistance, including KRAS, NRAS, BRAF, EGFR, ERBB2, and MET were detected. In conclusion, over 80% of patients with stage IV colorectal cancer had detectable ctDNA, and the majority had potentially actionable alterations. Concordance between tissue and blood was between 62% and 87%, despite a median of 5 months between tests. Resistance alterations emerged on anti-EGFR therapy. Therefore, biopsy-free, noninvasive ctDNA analysis provides data relevant to the clinical setting. Importantly, sequential ctDNA analysis detects patterns of emerging resistance allowing for precision planning of future therapy.
- Published
- 2019
10. Molecular profiling of cancer patients enables personalized combination therapy: the I-PREDICT study
- Author
-
Sicklick, Jason K, Kato, Shumei, Okamura, Ryosuke, Schwaederle, Maria, Hahn, Michael E, Williams, Casey B, De, Pradip, Krie, Amy, Piccioni, David E, Miller, Vincent A, Ross, Jeffrey S, Benson, Adam, Webster, Jennifer, Stephens, Philip J, Lee, J Jack, Fanta, Paul T, Lippman, Scott M, Leyland-Jones, Brian, and Kurzrock, Razelle
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Human Genome ,Cancer ,Clinical Trials and Supportive Activities ,Orphan Drug ,Cancer Genomics ,Clinical Research ,Women's Health ,Precision Medicine ,Health Disparities ,Rare Diseases ,Genetics ,6.1 Pharmaceuticals ,5.1 Pharmaceuticals ,Good Health and Well Being ,Adult ,Aged ,Aged ,80 and over ,Combined Modality Therapy ,Female ,Gene Expression Profiling ,High-Throughput Nucleotide Sequencing ,Humans ,Male ,Middle Aged ,Molecular Targeted Therapy ,Neoplasms ,Progression-Free Survival ,Prospective Studies ,Young Adult ,Medical and Health Sciences ,Immunology ,Biomedical and clinical sciences ,Health sciences - Abstract
Cancer treatments have evolved from indiscriminate cytotoxic agents to selective genome- and immune-targeted drugs that have transformed the outcomes of some malignancies1. Tumor complexity and heterogeneity suggest that the 'precision medicine' paradigm of cancer therapy requires treatment to be personalized to the individual patient2-6. To date, precision oncology trials have been based on molecular matching with predetermined monotherapies7-14. Several of these trials have been hindered by very low matching rates, often in the 5-10% range15, and low response rates. Low matching rates may be due to the use of limited gene panels, restrictive molecular matching algorithms, lack of drug availability, or the deterioration and death of end-stage patients before therapy can be implemented. We hypothesized that personalized treatment with combination therapies would improve outcomes in patients with refractory malignancies. As a first test of this concept, we implemented a cross-institutional prospective study (I-PREDICT, NCT02534675 ) that used tumor DNA sequencing and timely recommendations for individualized treatment with combination therapies. We found that administration of customized multidrug regimens was feasible, with 49% of consented patients receiving personalized treatment. Targeting of a larger fraction of identified molecular alterations, yielding a higher 'matching score', was correlated with significantly improved disease control rates, as well as longer progression-free and overall survival rates, compared to targeting of fewer somatic alterations. Our findings suggest that the current clinical trial paradigm for precision oncology, which pairs one driver mutation with one drug, may be optimized by treating molecularly complex and heterogeneous cancers with combinations of customized agents.
- Published
- 2019
11. Current management of succinate dehydrogenase deficient gastrointestinal stromal tumors
- Author
-
Neppala, Pushpa, Banerjee, Sudeep, Fanta, Paul T., Yerba, Mayra, Porras, Kevin A., Burgoyne, Adam M., and Sicklick, Jason K.
- Abstract
Gastrointestinal stromal tumors (GIST) are increasingly recognized as having diverse biology. With the development of tyrosine kinase inhibitors molecularly matched to oncogenic KIT and PDGFRA mutations, GIST have become a quintessential model for precision oncology. However, about 5-10% of GIST lack these driver mutations and are deficient in succinate dehydrogenase (SDH), an enzyme that converts succinate to fumarate. SDH deficiency leads to accumulation of succinate, an oncometabolite that promotes tumorigenesis. SDH- deficient GIST are clinically unique in that they generally affect younger patients and are associated with GIST- paraganglioma hereditary syndrome, also known as Carney-Stratakis Syndrome. SDH-deficient GIST are generally resistant to tyrosine-kinase inhibitors, the standard treatment for advanced or metastatic GIST. Thus, surgical resection is the mainstay of treatment for localized disease, but recurrence is common. Clinical trials are currently underway investigating systemic agents for treatment of advanced SDH-deficient GIST. However, further studies are warranted to improve our understanding of SDH-deficient GIST disease biology, natural history, surgical approaches, and novel therapeutics.
- Published
- 2019
12. Analysis of Circulating Tumor DNA and Clinical Correlates in Patients with Esophageal, Gastroesophageal Junction, and Gastric Adenocarcinoma
- Author
-
Kato, Shumei, Okamura, Ryosuke, Baumgartner, Joel M, Patel, Hitendra, Leichman, Lawrence, Kelly, Kaitlyn, Sicklick, Jason K, Fanta, Paul T, Lippman, Scott M, and Kurzrock, Razelle
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Oncology and Carcinogenesis ,Cancer ,Rare Diseases ,Genetics ,Digestive Diseases ,Human Genome ,Orphan Drug ,Genetic Testing ,Cancer Genomics ,Biotechnology ,Good Health and Well Being ,Adenocarcinoma ,Adult ,Aged ,Aged ,80 and over ,Biomarkers ,Tumor ,Circulating Tumor DNA ,Esophageal Neoplasms ,Esophagogastric Junction ,Female ,Gene Expression Profiling ,High-Throughput Nucleotide Sequencing ,Humans ,Kaplan-Meier Estimate ,Male ,Middle Aged ,Neoplasm Grading ,Neoplasm Staging ,Prognosis ,Stomach Neoplasms ,Treatment Outcome ,Young Adult ,Oncology & Carcinogenesis ,Clinical sciences ,Oncology and carcinogenesis - Abstract
PurposeEsophageal, gastroesophageal junction, and gastric adenocarcinoma (herein gastroesophageal adenocarcinomas) are associated with poor prognosis and limited systemic treatment options. To further understand the genomic landscape of gastroesophageal cancers and its clinical correlations, circulating tumor DNA (ctDNA) from patients' plasma was evaluated using next-generation sequencing (NGS).Experimental designWe analyzed genomic alterations of 55 patients (mostly advanced disease; 9, surgically resectable) with gastroesophageal adenocarcinomas using clinical-grade NGS performed on plasma-derived ctDNA (54-73 gene panel). The test detects single-nucleotide variants, as well as copy number amplifications, fusions, and indels in selected genes.ResultsSeventy-six percent of patients (42/55) had ≥1 genomic alteration [including variants of unknown significance (VUS)] and 69.1% (38/55) had ≥1 characterized alteration (excluding VUSs). The median number of alterations per patient was 2 (range, 0-15). TP53 (50.9%, 28/55), PIK3CA (16.4%, 9/55), ERBB2 (14.5%, 8/55), and KRAS (14.5%, 8/55) genes were most frequently affected characterized alterations. Thirty-one patients also had tissue NGS. Concordance between tissue and ctDNA ranged from 61.3% (TP53 alterations) to 87.1% (KRAS alterations). ERBB2 alterations were significantly associated with poor overall survival (HR, 14.06; 95% confidence interval, 2.44-81.03; P = 0.003 multivariate analysis). Among patients with ≥1 alteration, no 2 patients had identical molecular portfolios. All patients with ≥1 characterized alteration had theoretically targetable alterations by an FDA-approved agent (on- or off-label). Illustrative case treated with cognate agent is presented.ConclusionsEvaluation of ctDNA by NGS among patients with gastroesophageal adenocarcinoma is feasible. Patients harbored heterogeneous patterns of genomics, with most having alterations that are potentially pharmacologically tractable.
- Published
- 2018
13. Telomerase reverse transcriptase promoter alterations across cancer types as detected by next‐generation sequencing: A clinical and molecular analysis of 423 patients
- Author
-
Schwaederle, Maria, Krishnamurthy, Nithya, Daniels, Gregory A, Piccioni, David E, Kesari, Santosh, Fanta, Paul T, Schwab, Richard B, Patel, Sandip P, Parker, Barbara A, and Kurzrock, Razelle
- Subjects
Genetics ,Clinical Research ,Cancer ,2.1 Biological and endogenous factors ,Aetiology ,Biomarkers ,Tumor ,Female ,Follow-Up Studies ,Gene Expression Regulation ,Neoplastic ,High-Throughput Nucleotide Sequencing ,Humans ,Male ,Middle Aged ,Mutation ,Neoplasms ,Prognosis ,Promoter Regions ,Genetic ,Retrospective Studies ,Survival Rate ,Telomerase ,BRAF ,glioblastoma ,melanomas ,next-generation sequencing ,survival ,telomerase reverse transcriptase (TERT) promoter mutations ,Oncology and Carcinogenesis ,Public Health and Health Services ,Oncology & Carcinogenesis - Abstract
BACKGROUND:Telomerase reverse transcriptase (TERT) promoter mutations that may affect telomerase activity have recently been described in human malignancies. The purpose of this study was to investigate the clinical correlates of TERT promoter abnormalities in a large cohort of patients with diverse cancers. METHODS:This study analyzed TERT promoter alterations and clinical characteristics of 423 consecutive patients for whom molecular testing by next-generation sequencing was performed between August 2014 and July 2015. RESULTS:Of the 423 patients, 61 (14.4%) had TERT promoter mutations, and this placed TERT promoter alterations among the most prevalent aberrations after tumor protein 53 (TP53; 39%) and KRAS and cyclin-dependent kinase inhibitor 2A/B (CDKN2A/B) alterations (15% each) in this population. TERT promoter alterations were more frequent in men (P = .031) and were associated with brain cancers (P = .001), skin cancers/melanoma (P = .001), and a higher number of aberrations (P = .0001). A co-alteration analysis found that TERT promoter alterations were significantly correlated with CDKN2A/B (P = .001) and BRAF abnormalities (P = .0003). Patients harboring TERT promoter alterations or TP53 or CDKN2A/B alterations and those with 4 or more alterations demonstrated shorter survival (hazard ratio for normal TERT promoters vs aberrant ones, 0.44; P = .017). However, only a higher number of alterations remained significant in the multivariate analysis. CONCLUSIONS:Overall, TERT promoter alterations were among the most prevalent aberrations in this population, with very high rates in brain cancers (48% of patients) and melanomas (56% of patients). These aberrations frequently coexist with a high number of other aberrations, with the latter feature also significantly associated with poorer overall survival. Therapeutic options for targeting tumors with TERT promoter mutations are currently limited, although a variety of novel approaches are under development. Cancer 2018;124:1288-96. © 2017 American Cancer Society.
- Published
- 2018
14. The Mutational Landscape of Gastrointestinal Malignancies as Reflected by Circulating Tumor DNA
- Author
-
Riviere, Paul, Fanta, Paul T, Ikeda, Sadakatsu, Baumgartner, Joel, Heestand, Gregory M, and Kurzrock, Razelle
- Subjects
Rare Diseases ,Genetics ,Digestive Diseases ,Colo-Rectal Cancer ,Clinical Research ,Cancer ,Pancreatic Cancer ,Good Health and Well Being ,Adult ,Aged ,Aged ,80 and over ,Cell Line ,Tumor ,Circulating Tumor DNA ,Female ,Gastrointestinal Neoplasms ,Humans ,Male ,Middle Aged ,Mutation ,Retrospective Studies ,Oncology and Carcinogenesis ,Pharmacology and Pharmaceutical Sciences ,Oncology & Carcinogenesis - Abstract
We aimed to assess the utility of a novel, noninvasive method of detecting genomic alterations in patients with gastrointestinal malignancies, i.e., the use of liquid biopsies to obtain blood-derived circulating tumor DNA (ctDNA) through an analysis of the genomic landscape of ctDNA (68 genes) from 213 patients with advanced gastrointestinal cancers. The most common cancer types were colorectal adenocarcinoma (N = 55; 26%), appendiceal adenocarcinoma (N = 46; 22%), hepatocellular carcinoma (N = 31; 15%), and pancreatic ductal adenocarcinoma (N = 25; 12%). The majority of patients (58%) had ≥1 characterized alteration (excluded variants of unknown significance). The median number of characterized alterations was 1 (range, 0-13). The number of detected alterations per patient varied between different cancer types: in hepatocellular carcinoma, 74% of patients (23/31) had ≥1 characterized alteration(s) versus 24% of appendiceal adenocarcinoma patients (11/46). The median percent ctDNA among characterized alterations was 2.50% (interquartile range, 0.76%-8.96%). Overall, 95% of patients (117/123) had distinct molecular portfolios with 143 unique characterized alterations within 56 genes. Overall, concordance rates of 96%, 94%, 95%, and 91%, respectively, were found between ctDNA and tissue biopsy (N = 105 patients) in the four most common alterations (KRAS amplification, MYC amplification, KRAS G12V, and EGFR amplification). Of 123 patients with characterized alterations, >99% (122/123; 57% of entire population tested; 122/213) had one or more alterations potentially actionable by experimental or approved drugs. These observations suggest that many patients with gastrointestinal tumors, including difficult-to-biopsy malignancies like hepatocellular cancers, frequently have discernible and theoretically pharmacologically tractable ctDNA alterations that merit further studies in prospective trials. Mol Cancer Ther; 17(1); 297-305. ©2017 AACR.
- Published
- 2018
15. Next-generation sequencing in the clinical setting clarifies patient characteristics and potential actionability
- Author
-
Bieg-Bourne, Cheyennedra C, Millis, Sherri Z, Piccioni, David E, Fanta, Paul T, Goldberg, Michael E, Chmielecki, Juliann, Parker, Barbara A, and Kurzrock, Razelle
- Subjects
Clinical Research ,Cancer ,Biotechnology ,Genetics ,Good Health and Well Being ,Biomarkers ,Tumor ,DNA Mutational Analysis ,Genetic Predisposition to Disease ,High-Throughput Nucleotide Sequencing ,Humans ,Mutation ,Neoplasms ,Precision Medicine ,Reproducibility of Results ,Oncology and Carcinogenesis ,Oncology & Carcinogenesis - Abstract
Enhancements in clinical-grade next-generation sequencing (NGS) have fueled the advancement of precision medicine in the clinical oncology field. Here, we survey the molecular profiles of 1,113 patients with diverse malignancies who successfully underwent clinical-grade NGS (236-404 genes) in an academic tertiary cancer center. Among the individual tumors examined, the majority showed at least one detectable alteration (97.2%). Among 2,045 molecular aberrations was the involvement of 302 distinct genes. The most commonly altered genes were TP53 (47.0%), CDKN2A (18.0%), TERT (17.0%), and KRAS (16.0%), and the majority of patients had tumors that harbored multiple alterations. Tumors displayed a median of four alterations (range, 0-29). Most individuals had at least one potentially actionable alteration (94.7%), with the median number of potentially actionable alterations per patient being 2 (range, 0-13). A total of 1,048 (94.2%) patients exhibited a unique molecular profile, with either genes altered or loci within the gene(s) altered being distinct. Approximately 13% of patients displayed a genomic profile identical to at least one other patient; although genes altered were the same, the affected loci may have differed. Overall, our results underscore the complex heterogeneity of malignancies and argue that customized combination therapies will be essential to optimize cancer treatment regimens. Cancer Res; 77(22); 6313-20. ©2017 AACR.
- Published
- 2017
16. Duodenal-Jejunal Flexure GI Stromal Tumor Frequently Heralds Somatic NF1 and Notch Pathway Mutations
- Author
-
Burgoyne, Adam M, De Siena, Martina, Alkhuziem, Maha, Tang, Chih-Min, Medina, Benjamin, Fanta, Paul T, Belinsky, Martin G, von Mehren, Margaret, Thorson, John A, Madlensky, Lisa, Bowler, Timothy, D'Angelo, Francesco, Stupack, Dwayne G, Harismendy, Olivier, DeMatteo, Ronald P, and Sicklick, Jason K
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Human Genome ,Cancer Genomics ,Cancer ,Neurofibromatosis ,Genetics ,Digestive Diseases ,Rare Diseases ,Biotechnology ,Neurosciences ,2.1 Biological and endogenous factors ,Oncology and carcinogenesis - Abstract
PurposeGI stromal tumors (GISTs) are commonly associated with somatic mutations in KIT and PDGFRA. However, a subset arises from mutations in NF1, most commonly associated with neurofibromatosis type 1. We define the anatomic distribution of NF1 alterations in GIST.MethodsWe describe the demographic/clinicopathologic features of 177 patients from two institutions whose GISTs underwent next-generation sequencing of ≥315 cancer-related genes.ResultsWe initially identified six (9.7%) of 62 GISTs with NF1 genomic alterations from the first cohort. Of these six patients, five (83.3%) had unifocal tumors at the duodenal-jejunal flexure (DJF). Two additional patients with DJF GISTs had non-NF1 (KIT and BRAF) genomic alterations. After excluding one DJF GIST with an NF1 single nucleotide polymorphism, four (57.1%) of seven sequenced DJF tumors demonstrated deleterious NF1 alterations, whereas only one (1.8%) of 55 sequenced non-DJF GISTs had a deleterious NF1 somatic mutation (P < .001). One patient with DJF GIST had a germline NF1 variant that was associated with incomplete penetrance of clinical neurofibromatosis type 1 features along with a somatic NF1 mutation. Of the five DJF GISTs with any NF1 alteration, three (60%) had KIT mutations, and three (60%) had Notch pathway mutations (NOTCH2, MAML2, CDC73). We validated these findings in a second cohort of 115 GISTs, where two (40%) of five unifocal NF1-mutated GISTs arose at the DJF, and one of these also had a Notch pathway mutation (EP300).ConclusionBroad genomic profiling of adult GISTs has revealed that NF1 alterations are enriched in DJF GISTs. These tumors also may harbor concurrent activating KIT and/or inactivating Notch pathway mutations. In some cases, germline NF1 genetic testing may be appropriate for patients with DJF GISTs.
- Published
- 2017
17. Genomic Alterations in Circulating Tumor DNA from Diverse Cancer Patients Identified by Next-Generation Sequencing
- Author
-
Schwaederle, Maria, Chattopadhyay, Ranajoy, Kato, Shumei, Fanta, Paul T, Banks, Kimberly C, Choi, In Sil, Piccioni, David E, Ikeda, Sadakatsu, Talasaz, AmirAli, Lanman, Richard B, Bazhenova, Lyudmila, and Kurzrock, Razelle
- Subjects
Cancer ,Biotechnology ,Human Genome ,Clinical Research ,Genetics ,Rare Diseases ,Good Health and Well Being ,Adolescent ,Adult ,Aged ,Aged ,80 and over ,Biomarkers ,Tumor ,Child ,Child ,Preschool ,DNA ,Neoplasm ,Female ,Genomics ,High-Throughput Nucleotide Sequencing ,Humans ,Male ,Middle Aged ,Mutation ,Neoplasms ,Retrospective Studies ,Young Adult ,Oncology and Carcinogenesis ,Oncology & Carcinogenesis - Abstract
Noninvasive genomic profiling of tumors may be possible with next-generation sequencing (NGS) of blood-derived circulating tumor DNA (ctDNA), but proof of concept in a large cohort of patients with diverse cancers has yet to be reported. Here we report the results of an analysis of plasma-derived ctDNA from 670 patients with diverse cancers. The tumors represented in the patient cohort were mainly gastrointestinal (31.8%), brain (22.7%), or lung (20.7%). ctDNA obtained from most patients [N = 423 (63%)] displayed at least one alteration. The most frequent alterations seen, as characterized mutations or variants of unknown significance, occurred in TP53 (32.5% of patients), EGFR (13%), KRAS (12.5%), and PIK3CA (9.1%); for characterized alterations, 30.7% (TP53), 7.6% (EGFR), 12.2% (KRAS), and 7.7% (PIK3CA). We found that 32% of brain tumors had at least one ctDNA alteration. Head and neck tumors were independently associated with a higher number of alterations in a multivariable analysis (P = 0.019). Notably, 320/670 (48%) of patients displayed potentially actionable alterations, with 241 patients possible candidates for on-label or off-label treatment with an FDA-approved drug. Several illustrations of the clinical utility of the information obtained for improving treatment of specific patients is provided. Our findings demonstrate the feasibility and impact of genomic profiling of tumors by ctDNA NGS, greatly encouraging broader investigations of the application of this technology for precision medicine in cancer management. Cancer Res; 77(19); 5419-27. ©2017 AACR.
- Published
- 2017
18. FGFR1 and NTRK3 actionable alterations in "Wild-Type" gastrointestinal stromal tumors.
- Author
-
Shi, Eileen, Chmielecki, Juliann, Tang, Chih-Min, Wang, Kai, Heinrich, Michael C, Kang, Guhyun, Corless, Christopher L, Hong, David, Fero, Katherine E, Murphy, James D, Fanta, Paul T, Ali, Siraj M, De Siena, Martina, Burgoyne, Adam M, Movva, Sujana, Madlensky, Lisa, Heestand, Gregory M, Trent, Jonathan C, Kurzrock, Razelle, Morosini, Deborah, Ross, Jeffrey S, Harismendy, Olivier, and Sicklick, Jason K
- Subjects
Humans ,Gastrointestinal Stromal Tumors ,Receptor ,trkC ,Oncogene Proteins ,Fusion ,Demography ,Mutation ,Genome ,Human ,Adult ,Female ,Male ,Receptor ,Fibroblast Growth Factor ,Type 1 ,ETV6–NTRK3 ,FGFR1 ,GIST ,Gene sequencing ,ETV6-NTRK3 ,Digestive Diseases ,Biotechnology ,Cancer ,Genetics ,2.1 Biological and endogenous factors ,Immunology ,Medical and Health Sciences - Abstract
BackgroundAbout 10-15% of adult, and most pediatric, gastrointestinal stromal tumors (GIST) lack mutations in KIT, PDGFRA, SDHx, or RAS pathway components (KRAS, BRAF, NF1). The identification of additional mutated genes in this rare subset of tumors can have important clinical benefit to identify altered biological pathways and select targeted therapies.MethodsWe performed comprehensive genomic profiling (CGP) for coding regions in more than 300 cancer-related genes of 186 GISTs to assess for their somatic alterations.ResultsWe identified 24 GIST lacking alterations in the canonical KIT/PDGFRA/RAS pathways, including 12 without SDHx alterations. These 24 patients were mostly adults (96%). The tumors had a 46% rate of nodal metastases. These 24 GIST were more commonly mutated at 7 genes: ARID1B, ATR, FGFR1, LTK, SUFU, PARK2 and ZNF217. Two tumors harbored FGFR1 gene fusions (FGFR1-HOOK3, FGFR1-TACC1) and one harbored an ETV6-NTRK3 fusion that responded to TRK inhibition. In an independent sample set, we identified 5 GIST cases lacking alterations in the KIT/PDGFRA/SDHx/RAS pathways, including two additional cases with FGFR1-TACC1 and ETV6-NTRK3 fusions.ConclusionsUsing patient demographics, tumor characteristics, and CGP, we show that GIST lacking alterations in canonical genes occur in younger patients, frequently metastasize to lymph nodes, and most contain deleterious genomic alterations, including gene fusions involving FGFR1 and NTRK3. If confirmed in larger series, routine testing for these translocations may be indicated for this subset of GIST. Moreover, these findings can be used to guide personalized treatments for patients with GIST. Trial registration NCT 02576431. Registered October 12, 2015.
- Published
- 2016
19. Use of Liquid Biopsies in Clinical Oncology: Pilot Experience in 168 Patients
- Author
-
Schwaederle, Maria, Husain, Hatim, Fanta, Paul T, Piccioni, David E, Kesari, Santosh, Schwab, Richard B, Patel, Sandip P, Harismendy, Olivier, Ikeda, Megumi, Parker, Barbara A, and Kurzrock, Razelle
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Oncology and Carcinogenesis ,Cancer ,Clinical Research ,Good Health and Well Being ,Circulating Tumor DNA ,DNA ,Neoplasm ,ErbB Receptors ,Female ,Genomics ,High-Throughput Nucleotide Sequencing ,Humans ,Liquid Biopsy ,Male ,Medical Oncology ,Middle Aged ,Neoplasms ,Phosphatidylinositol 3-Kinases ,Receptor ,ErbB-2 ,Retrospective Studies ,Tumor Suppressor Protein p53 ,Receptor ,erbB-2 ,Oncology & Carcinogenesis ,Clinical sciences ,Oncology and carcinogenesis - Abstract
PurposeThere is a growing interest in using circulating tumor DNA (ctDNA) testing in patients with cancer.Experimental designA total of 168 patients with diverse cancers were analyzed. Patients had digital next-generation sequencing (54 cancer-related gene panel including amplifications in ERBB2, EGFR, and MET) performed on their plasma. Type of genomic alterations, potential actionability, concordance with tissue testing, and patient outcome were examined.ResultsFifty-eight percent of patients (98/168) had ≥1 ctDNA alteration(s). Of the 98 patients with alterations, 71.4% had ≥ 1 alteration potentially actionable by an FDA-approved drug. The median time interval between the tissue biopsy and the blood draw was 2.7 months for patients with ≥ 1 alteration in common compared with 14.4 months (P = 0.006) for the patients in whom no common alterations were identified in the tissue and plasma. Overall concordance rates for tissue and ctDNA were 70.3% for TP53 and EGFR, 88.1% for PIK3CA, and 93.1% for ERBB2 alterations. There was a significant correlation between the cases with ≥ 1 alteration with ctDNA ≥ 5% and shorter survival (median = 4.03 months vs. not reached at median follow-up of 6.1 months; P < 0.001). Finally, 5 of the 12 evaluable patients (42%) matched to a treatment targeting an alteration(s) detected in their ctDNA test achieved stable disease ≥ 6 months/partial remission compared with 2 of 28 patients (7.1%) for the unmatched patients, P = 0.02.ConclusionsOur initial study demonstrates that ctDNA tests provide information complementary to that in tissue biopsies and may be useful in determining prognosis and treatment. Clin Cancer Res; 22(22); 5497-505. ©2016 AACR.
- Published
- 2016
20. Genomics of gallbladder cancer: the case for biomarker-driven clinical trial design
- Author
-
Sicklick, Jason K, Fanta, Paul T, Shimabukuro, Kelly, and Kurzrock, Razelle
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Immunology ,Genetics ,Human Genome ,Biotechnology ,Rare Diseases ,Digestive Diseases ,Cancer ,Good Health and Well Being ,Antineoplastic Combined Chemotherapy Protocols ,Biliary Tract Neoplasms ,Biomarkers ,Tumor ,Carcinoma ,Clinical Trials as Topic ,Disease Management ,Gallbladder Neoplasms ,Gene Expression Regulation ,Neoplastic ,Genetic Predisposition to Disease ,Genetic Variation ,Genomics ,Humans ,Molecular Targeted Therapy ,Precision Medicine ,Prognosis ,Research Design ,Signal Transduction ,Standard of Care ,Treatment Outcome ,Biliary tract cancers ,Cholangiocarcinoma ,Gallbladder cancer ,Targeted therapy ,Oncology & Carcinogenesis ,Oncology and carcinogenesis - Abstract
Background and aimsGallbladder carcinoma is a rare, aggressive malignancy of the biliary tract associated with a poor prognosis. Despite the deployment of targeted therapies that have demonstrated marked survival benefits in many tumor types, traditional cytotoxic chemotherapy has remained the mainstay of treatment for unresectable and metastatic gallbladder cancer.MethodsSystematic review of ongoing and prior clinical studies shows a paucity of biomarker-driven therapeutic trials using targeted agents in gallbladder cancer. In fact, over the past 6 years, of the 38 therapeutic biliary tract protocols listed on clinicaltrials.gov, only 6 (21 %) utilized targeted therapies based upon tumor biomarkers or genomics. Now that we have entered the era of next-generation sequencing and precision medicine, we are beginning to identify common and specific genetic alterations in gallbladder carcinomas.ResultsA review of the literature reveals alterations in ARID1A, BRAF, CDKN2A/B, EGFR, ERBB2-4, HKN-RAS, PIK3CA, PBRM1, and TP53. Given the widespread use of tumor genomic profiling and the fact that most of the aforementioned alterations are pharmacologically tractable, these observations suggest the potential for new therapeutic strategies in this aggressive malignancy.ConclusionsTaken together, further understanding of the genomic landscape of gallbladder cancer coupled with biomarker-driven clinical trials that match therapies to targets are urgently needed.
- Published
- 2016
21. Precision Oncology: The UC San Diego Moores Cancer Center PREDICT Experience
- Author
-
Schwaederle, Maria, Parker, Barbara A, Schwab, Richard B, Daniels, Gregory A, Piccioni, David E, Kesari, Santosh, Helsten, Teresa L, Bazhenova, Lyudmila A, Romero, Julio, Fanta, Paul T, Lippman, Scott M, and Kurzrock, Razelle
- Subjects
Oncology & Carcinogenesis ,Oncology and Carcinogenesis ,Pharmacology and Pharmaceutical Sciences - Published
- 2016
22. Detection rate of actionable mutations in diverse cancers using a biopsy-free (blood) circulating tumor cell DNA assay.
- Author
-
Schwaederle, Maria, Husain, Hatim, Fanta, Paul T, Piccioni, David E, Kesari, Santosh, Schwab, Richard B, Banks, Kimberly C, Lanman, Richard B, Talasaz, AmirAli, Parker, Barbara A, and Kurzrock, Razelle
- Subjects
Humans ,Neoplasms ,DNA ,Neoplasm ,Biopsy ,Retrospective Studies ,Mutation ,Adult ,Aged ,Aged ,80 and over ,Middle Aged ,Female ,Male ,Tumor Suppressor Protein p53 ,Proto-Oncogene Proteins c-met ,Receptor ,Notch1 ,Neoplastic Cells ,Circulating ,Young Adult ,DNA Copy Number Variations ,Phosphatidylinositol 3-Kinases ,Class I Phosphatidylinositol 3-Kinases ,High-Throughput Nucleotide Sequencing ,ErbB Receptors ,Biomarkers ,Tumor ,actionable alteration ,cancer ,ctDNA ,liquid biopsy ,personalized therapy ,Clinical Research ,Human Genome ,Rare Diseases ,Cancer ,Genetics ,Brain Disorders ,Biotechnology ,Brain Cancer ,Oncology and Carcinogenesis - Abstract
Analysis of cell-free DNA using next-generation sequencing (NGS) is a powerful tool for the detection/monitoring of alterations present in circulating tumor DNA (ctDNA). Plasma extracted from 171 patients with a variety of cancers was analyzed for ctDNA (54 genes and copy number variants (CNVs) in three genes (EGFR, ERBB2 and MET)). The most represented cancers were lung (23%), breast (23%), and glioblastoma (19%). Ninety-nine patients (58%) had at least one detectable alteration. The most frequent alterations were TP53 (29.8%), followed by EGFR (17.5%), MET (10.5%), PIK3CA (7%), and NOTCH1 (5.8%). In contrast, of 222 healthy volunteers, only one had an aberration (TP53). Ninety patients with non-brain tumors had a discernible aberration (65% of 138 patients; in 70% of non-brain tumor patients with an alteration, the anomaly was potentially actionable). Interestingly, nine of 33 patients (27%) with glioblastoma had an alteration (6/33 (18%) potentially actionable). Overall, sixty-nine patients had potentially actionable alterations (40% of total; 69.7% of patients (69/99) with alterations); 68 patients (40% of total; 69% of patients with alterations), by a Food and Drug Administration (FDA) approved drug. In summary, 65% of diverse cancers (as well as 27% of glioblastomas) had detectable ctDNA aberration(s), with the majority theoretically actionable by an approved agent.
- Published
- 2016
23. Molecular inimitability amongst tumors: implications for precision cancer medicine in the age of personalized oncology
- Author
-
Patel, Sandip P, Schwaederle, Maria, Daniels, Gregory A, Fanta, Paul T, Schwab, Richard B, Shimabukuro, Kelly A, Kesari, Santosh, Piccioni, David E, Bazhenova, Lyudmila A, Helsten, Teresa L, Lippman, Scott M, Parker, Barbara A, and Kurzrock, Razelle
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Oncology and Carcinogenesis ,Rare Diseases ,Brain Disorders ,Brain Cancer ,Lung ,Lung Cancer ,Cancer ,Genetics ,Good Health and Well Being ,Biomarkers ,Tumor ,Computational Biology ,DNA Mutational Analysis ,Databases ,Genetic ,Female ,Gene Expression Profiling ,Genetic Predisposition to Disease ,High-Throughput Nucleotide Sequencing ,Humans ,Male ,Middle Aged ,Mutation ,Neoplasms ,Patient Selection ,Phenotype ,Precision Medicine ,Predictive Value of Tests ,Prognosis ,personalized medicine ,genomics ,cancer ,next-generation sequencing ,clinical trials ,Oncology and carcinogenesis - Abstract
Tumor sequencing has revolutionized oncology, allowing for detailed interrogation of the molecular underpinnings of cancer at an individual level. With this additional insight, it is increasingly apparent that not only do tumors vary within a sample (tumor heterogeneity), but also that each patient's individual tumor is a constellation of unique molecular aberrations that will require an equally unique personalized therapeutic regimen. We report here the results of 439 patients who underwent Clinical Laboratory Improvement Amendment (CLIA)-certified next generation sequencing (NGS) across histologies. Among these patients, 98.4% had a unique molecular profile, and aside from three primary brain tumor patients with a single genetic lesion (IDH1 R132H), no two patients within a given histology were molecularly identical. Additionally, two sets of patients had identical profiles consisting of two mutations in common and no other anomalies. However, these profiles did not segregate by histology (lung adenocarcinoma-appendiceal cancer (KRAS G12D and GNAS R201C), and lung adenocarcinoma-liposarcoma (CDK4 and MDM2 amplification pairs)). These findings suggest that most advanced tumors are molecular singletons within and between histologies, and that tumors that differ in histology may still nonetheless exhibit identical molecular portraits, albeit rarely.
- Published
- 2015
24. Next generation sequencing demonstrates association between tumor suppressor gene aberrations and poor outcome in patients with cancer
- Author
-
Schwaederle, Maria, Daniels, Gregory A, Piccioni, David E, Kesari, Santosh, Fanta, Paul T, Schwab, Richard B, Shimabukuro, Kelly A, Parker, Barbara A, and Kurzrock, Razelle
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Digestive Diseases ,Cancer ,2.1 Biological and endogenous factors ,Aetiology ,Good Health and Well Being ,Adenomatous Polyposis Coli Protein ,Female ,Genes ,Tumor Suppressor ,High-Throughput Nucleotide Sequencing ,Humans ,Kaplan-Meier Estimate ,Male ,Middle Aged ,Neoplasm Metastasis ,Neoplasms ,PTEN Phosphohydrolase ,Patient Outcome Assessment ,Precision Medicine ,Regression Analysis ,Time Factors ,Tumor Suppressor Protein p53 ,Tumor Suppressor Proteins ,tumor suppressor ,next-generation sequencing ,PTEN ,patient's outcome ,APC ,TP53 ,CDKN2A ,cancer ,Developmental Biology ,Biochemistry and cell biology - Abstract
Next generation sequencing is transforming patient care by allowing physicians to customize and match treatment to their patients' tumor alterations. Our goal was to study the association between key molecular alterations and outcome parameters. We evaluated the characteristics and outcomes (overall survival (OS), time to metastasis/recurrence, and best progression-free survival (PFS)) of 392 patients for whom next generation sequencing (182 or 236 genes) had been performed. The Kaplan-Meier method and Cox regression models were used for our analysis, and results were subjected to internal validation using a resampling method (bootstrap analysis). In a multivariable analysis (Cox regression model), the parameters that were statistically associated with a poorer overall survival were the presence of metastases at diagnosis (P = 0.014), gastrointestinal histology (P < 0.0001), PTEN (P < 0.0001), and CDKN2A alterations (P = 0.0001). The variables associated with a shorter time to metastases/recurrence were gastrointestinal histology (P = 0.004), APC (P = 0.008), PTEN (P = 0.026) and TP53 (P = 0.044) alterations. TP53 (P = 0.003) and PTEN (P = 0.034) alterations were independent predictors of a shorter best PFS. A personalized treatment approach (matching the molecular aberration with a cognate targeted drug) also correlated with a longer best PFS (P = 0.046). Our study demonstrated that, across diverse cancers, anomalies in specific tumor suppressor genes (PTEN, CDKN2A, APC, and/or TP53) were independently associated with a worse outcome, as reflected by time to metastases/recurrence, best PFS on treatment, and/or overall survival. These observations suggest that molecular diagnostic tests may provide important prognostic information in patients with cancer.
- Published
- 2015
25. Molecular profiling of advanced malignancies guides first-line N-of-1 treatments in the I-PREDICT treatment-naïve study
- Author
-
Sicklick, Jason K., Kato, Shumei, Okamura, Ryosuke, Patel, Hitendra, Nikanjam, Mina, Fanta, Paul T., Hahn, Michael E., De, Pradip, Williams, Casey, Guido, Jessica, Solomon, Benjamin M., McKay, Rana R., Krie, Amy, Boles, Sarah G., Ross, Jeffrey S., Lee, J. Jack, Leyland-Jones, Brian, Lippman, Scott M., and Kurzrock, Razelle
- Published
- 2021
- Full Text
- View/download PDF
26. Cyclin alterations in diverse cancers: Outcome and co-amplification network
- Author
-
Schwaederlé, Maria, Daniels, Gregory A, Piccioni, David E, Fanta, Paul T, Schwab, Richard B, Shimabukuro, Kelly A, Parker, Barbara A, and Kurzrock, Razelle
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Cancer ,Digestive Diseases ,Biomarkers ,Tumor ,Cyclins ,Disease-Free Survival ,Female ,Gene Amplification ,Gene Expression Regulation ,Neoplastic ,Gene Regulatory Networks ,Genetic Predisposition to Disease ,Humans ,Kaplan-Meier Estimate ,Logistic Models ,Male ,Middle Aged ,Multivariate Analysis ,Neoplasm Metastasis ,Neoplasm Recurrence ,Local ,Neoplasms ,Odds Ratio ,Phenotype ,Proportional Hazards Models ,Retrospective Studies ,Risk Factors ,Time Factors ,cyclin ,next generation sequencing ,molecular profile ,amplification ,Oncology and carcinogenesis - Abstract
Cyclin genes are key regulatory components of the cell cycle. With the development of new agents, cyclin-related genes are becoming increasingly important as they can be targeted. Yet, the biological implications of these alterations have not been fully studied. Clinical characteristics and outcome parameters were compared for patients harboring cyclin alterations versus not. CCN alterations were found in 13% of our population (50/392; all amplifications) and were associated with breast cancer (P < 0.0001), a higher median number of concomitant molecular alterations (P < 0.0001), and liver metastases (P = 0.046). Harboring a cyclin amplification was not associated with overall survival, the time to metastasis/recurrence, nor with the best progression-free survival. In a Cox regression model, gastrointestinal histology (P < 0.0001), PTEN (P < 0.0001), and CDK alterations (P = 0.041) had a significant association with poorer overall survival. CCN amplifications significantly correlated with alterations in FGF/FGFR family genes as well as in MET and ARFRP1. An extended correlation study shed light on a network of co-amplifications influenced in part by genes that were localized on the same amplicons. CCN amplifications are common across cancers and had distinctive biological associations. Customized combinations targeting the cyclin pathway as well as the extended co- amplification network may be necessary in order to address resistance mechanisms.
- Published
- 2015
27. Molecular Tumor Board: The University of California San Diego Moores Cancer Center Experience
- Author
-
Schwaederle, Maria, Parker, Barbara A, Schwab, Richard B, Fanta, Paul T, Boles, Sarah G, Daniels, Gregory A, Bazhenova, Lyudmila A, Subramanian, Rupa, Coutinho, Alice C, Ojeda‐Fournier, Haydee, Datnow, Brian, Webster, Nicholas J, Lippman, Scott M, and Kurzrock, Razelle
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Oncology and Carcinogenesis ,Clinical Research ,Biotechnology ,Precision Medicine ,Genetics ,Human Genome ,Cancer ,Good Health and Well Being ,Aged ,Disease-Free Survival ,Female ,Genome ,Human ,High-Throughput Nucleotide Sequencing ,Humans ,Male ,Middle Aged ,Neoplasms ,Pathology ,Molecular ,Treatment Outcome ,Molecular tumor board ,Molecular profile ,Personalized ,Mutation ,Oncology & Carcinogenesis ,Oncology and carcinogenesis - Abstract
ObjectiveDNA sequencing tests are enabling physicians to interrogate the molecular profiles of patients' tumors, but most oncologists have not been trained in advanced genomics. We initiated a molecular tumor board to provide expert multidisciplinary input for these patients.Materials and methodsA team that included clinicians, basic scientists, geneticists, and bioinformatics/pathway scientists with expertise in various cancer types attended. Molecular tests were performed in a Clinical Laboratory Improvement Amendments environment.ResultsPatients (n = 34, since December 2012) had received a median of three prior therapies. The median time from physician order to receipt of molecular diagnostic test results was 27 days (range: 14-77 days). Patients had a median of 4 molecular abnormalities (range: 1-14 abnormalities) found by next-generation sequencing (182- or 236-gene panels). Seventy-four genes were involved, with 123 distinct abnormalities. Importantly, no two patients had the same aberrations, and 107 distinct abnormalities were seen only once. Among the 11 evaluable patients whose treatment had been informed by molecular diagnostics, 3 achieved partial responses (progression-free survival of 3.4 months, ≥6.5 months, and 7.6 months). The most common reasons for being unable to act on the molecular diagnostic results were that patients were ineligible for or could not travel to an appropriately targeted clinical trial and/or that insurance would not cover the cognate agents.ConclusionGenomic sequencing is revealing complex molecular profiles that differ by patient. Multidisciplinary molecular tumor boards may help optimize management. Barriers to personalized therapy include access to appropriately targeted drugs.
- Published
- 2014
28. Population-Based Epidemiology and Mortality of Small Malignant Gastrointestinal Stromal Tumors in the USA
- Author
-
Coe, Taylor M., Fero, Katherine E., Fanta, Paul T., Mallory, Robert J., Tang, Chih-Min, Murphy, James D., and Sicklick, Jason K.
- Published
- 2016
- Full Text
- View/download PDF
29. Supplementary Table from Genomic Profiling of Blood-Derived Circulating Tumor DNA from Patients with Colorectal Cancer: Implications for Response and Resistance to Targeted Therapeutics
- Author
-
Choi, In Sil, primary, Kato, Shumei, primary, Fanta, Paul T., primary, Leichman, Lawrence, primary, Okamura, Ryosuke, primary, Raymond, Victoria M., primary, Lanman, Richard B., primary, Lippman, Scott M., primary, and Kurzrock, Razelle, primary
- Published
- 2023
- Full Text
- View/download PDF
30. Data from Genomic Profiling of Blood-Derived Circulating Tumor DNA from Patients with Colorectal Cancer: Implications for Response and Resistance to Targeted Therapeutics
- Author
-
Choi, In Sil, primary, Kato, Shumei, primary, Fanta, Paul T., primary, Leichman, Lawrence, primary, Okamura, Ryosuke, primary, Raymond, Victoria M., primary, Lanman, Richard B., primary, Lippman, Scott M., primary, and Kurzrock, Razelle, primary
- Published
- 2023
- Full Text
- View/download PDF
31. Supplementary Figure from Genomic Profiling of Blood-Derived Circulating Tumor DNA from Patients with Colorectal Cancer: Implications for Response and Resistance to Targeted Therapeutics
- Author
-
Choi, In Sil, primary, Kato, Shumei, primary, Fanta, Paul T., primary, Leichman, Lawrence, primary, Okamura, Ryosuke, primary, Raymond, Victoria M., primary, Lanman, Richard B., primary, Lippman, Scott M., primary, and Kurzrock, Razelle, primary
- Published
- 2023
- Full Text
- View/download PDF
32. Supplementary Table 1 from On the Road to Precision Cancer Medicine: Analysis of Genomic Biomarker Actionability in 439 Patients
- Author
-
Schwaederle, Maria, primary, Daniels, Gregory A., primary, Piccioni, David E., primary, Fanta, Paul T., primary, Schwab, Richard B., primary, Shimabukuro, Kelly A., primary, Parker, Barbara A., primary, and Kurzrock, Razelle, primary
- Published
- 2023
- Full Text
- View/download PDF
33. Data from The Mutational Landscape of Gastrointestinal Malignancies as Reflected by Circulating Tumor DNA
- Author
-
Riviere, Paul, primary, Fanta, Paul T., primary, Ikeda, Sadakatsu, primary, Baumgartner, Joel, primary, Heestand, Gregory M., primary, and Kurzrock, Razelle, primary
- Published
- 2023
- Full Text
- View/download PDF
34. Supplementary Tables 1-3, Supplementary Figure 1, and supplementary references from The Mutational Landscape of Gastrointestinal Malignancies as Reflected by Circulating Tumor DNA
- Author
-
Riviere, Paul, primary, Fanta, Paul T., primary, Ikeda, Sadakatsu, primary, Baumgartner, Joel, primary, Heestand, Gregory M., primary, and Kurzrock, Razelle, primary
- Published
- 2023
- Full Text
- View/download PDF
35. Data from On the Road to Precision Cancer Medicine: Analysis of Genomic Biomarker Actionability in 439 Patients
- Author
-
Schwaederle, Maria, primary, Daniels, Gregory A., primary, Piccioni, David E., primary, Fanta, Paul T., primary, Schwab, Richard B., primary, Shimabukuro, Kelly A., primary, Parker, Barbara A., primary, and Kurzrock, Razelle, primary
- Published
- 2023
- Full Text
- View/download PDF
36. Supplemental Methods; Supplemental Tables 1 and 2 from Precision Oncology: The UC San Diego Moores Cancer Center PREDICT Experience
- Author
-
Schwaederle, Maria, primary, Parker, Barbara A., primary, Schwab, Richard B., primary, Daniels, Gregory A., primary, Piccioni, David E., primary, Kesari, Santosh, primary, Helsten, Teresa L., primary, Bazhenova, Lyudmila A., primary, Romero, Julio, primary, Fanta, Paul T., primary, Lippman, Scott M., primary, and Kurzrock, Razelle, primary
- Published
- 2023
- Full Text
- View/download PDF
37. Data from Precision Oncology: The UC San Diego Moores Cancer Center PREDICT Experience
- Author
-
Schwaederle, Maria, primary, Parker, Barbara A., primary, Schwab, Richard B., primary, Daniels, Gregory A., primary, Piccioni, David E., primary, Kesari, Santosh, primary, Helsten, Teresa L., primary, Bazhenova, Lyudmila A., primary, Romero, Julio, primary, Fanta, Paul T., primary, Lippman, Scott M., primary, and Kurzrock, Razelle, primary
- Published
- 2023
- Full Text
- View/download PDF
38. Supplemental Table S3 from Next-Generation Sequencing in the Clinical Setting Clarifies Patient Characteristics and Potential Actionability
- Author
-
Bieg-Bourne, Cheyennedra C., primary, Millis, Sherri Z., primary, Piccioni, David E., primary, Fanta, Paul T., primary, Goldberg, Michael E., primary, Chmielecki, Juliann, primary, Parker, Barbara A., primary, and Kurzrock, Razelle, primary
- Published
- 2023
- Full Text
- View/download PDF
39. Supplementary Table S1 from Establishment of Patient-Derived Succinate Dehydrogenase–Deficient Gastrointestinal Stromal Tumor Models for Predicting Therapeutic Response
- Author
-
Yebra, Mayra, primary, Bhargava, Shruti, primary, Kumar, Avi, primary, Burgoyne, Adam M., primary, Tang, Chih-Min, primary, Yoon, Hyunho, primary, Banerjee, Sudeep, primary, Aguilera, Joseph, primary, Cordes, Thekla, primary, Sheth, Vipul, primary, Noh, Sangkyu, primary, Ustoy, Rowan, primary, Li, Sam, primary, Advani, Sunil J., primary, Corless, Christopher L., primary, Heinrich, Michael C., primary, Kurzrock, Razelle, primary, Lippman, Scott M., primary, Fanta, Paul T., primary, Harismendy, Olivier, primary, Metallo, Christian, primary, and Sicklick, Jason K., primary
- Published
- 2023
- Full Text
- View/download PDF
40. Supplementary Table 1 from Genomic Alterations in Circulating Tumor DNA from Diverse Cancer Patients Identified by Next-Generation Sequencing
- Author
-
Schwaederle, Maria, primary, Chattopadhyay, Ranajoy, primary, Kato, Shumei, primary, Fanta, Paul T., primary, Banks, Kimberly C., primary, Choi, In Sil, primary, Piccioni, David E., primary, Ikeda, Sadakatsu, primary, Talasaz, AmirAli, primary, Lanman, Richard B., primary, Bazhenova, Lyudmila, primary, and Kurzrock, Razelle, primary
- Published
- 2023
- Full Text
- View/download PDF
41. Supplementary Data from Analysis of Circulating Tumor DNA and Clinical Correlates in Patients with Esophageal, Gastroesophageal Junction, and Gastric Adenocarcinoma
- Author
-
Kato, Shumei, primary, Okamura, Ryosuke, primary, Baumgartner, Joel M., primary, Patel, Hitendra, primary, Leichman, Lawrence, primary, Kelly, Kaitlyn, primary, Sicklick, Jason K., primary, Fanta, Paul T., primary, Lippman, Scott M., primary, and Kurzrock, Razelle, primary
- Published
- 2023
- Full Text
- View/download PDF
42. Supplementary Figure S3 from Establishment of Patient-Derived Succinate Dehydrogenase–Deficient Gastrointestinal Stromal Tumor Models for Predicting Therapeutic Response
- Author
-
Yebra, Mayra, primary, Bhargava, Shruti, primary, Kumar, Avi, primary, Burgoyne, Adam M., primary, Tang, Chih-Min, primary, Yoon, Hyunho, primary, Banerjee, Sudeep, primary, Aguilera, Joseph, primary, Cordes, Thekla, primary, Sheth, Vipul, primary, Noh, Sangkyu, primary, Ustoy, Rowan, primary, Li, Sam, primary, Advani, Sunil J., primary, Corless, Christopher L., primary, Heinrich, Michael C., primary, Kurzrock, Razelle, primary, Lippman, Scott M., primary, Fanta, Paul T., primary, Harismendy, Olivier, primary, Metallo, Christian, primary, and Sicklick, Jason K., primary
- Published
- 2023
- Full Text
- View/download PDF
43. Data from Next-Generation Sequencing in the Clinical Setting Clarifies Patient Characteristics and Potential Actionability
- Author
-
Bieg-Bourne, Cheyennedra C., primary, Millis, Sherri Z., primary, Piccioni, David E., primary, Fanta, Paul T., primary, Goldberg, Michael E., primary, Chmielecki, Juliann, primary, Parker, Barbara A., primary, and Kurzrock, Razelle, primary
- Published
- 2023
- Full Text
- View/download PDF
44. Supplementary Data from Establishment of Patient-Derived Succinate Dehydrogenase–Deficient Gastrointestinal Stromal Tumor Models for Predicting Therapeutic Response
- Author
-
Yebra, Mayra, primary, Bhargava, Shruti, primary, Kumar, Avi, primary, Burgoyne, Adam M., primary, Tang, Chih-Min, primary, Yoon, Hyunho, primary, Banerjee, Sudeep, primary, Aguilera, Joseph, primary, Cordes, Thekla, primary, Sheth, Vipul, primary, Noh, Sangkyu, primary, Ustoy, Rowan, primary, Li, Sam, primary, Advani, Sunil J., primary, Corless, Christopher L., primary, Heinrich, Michael C., primary, Kurzrock, Razelle, primary, Lippman, Scott M., primary, Fanta, Paul T., primary, Harismendy, Olivier, primary, Metallo, Christian, primary, and Sicklick, Jason K., primary
- Published
- 2023
- Full Text
- View/download PDF
45. Supplemental material from Use of Liquid Biopsies in Clinical Oncology: Pilot Experience in 168 Patients
- Author
-
Schwaederle, Maria, primary, Husain, Hatim, primary, Fanta, Paul T., primary, Piccioni, David E., primary, Kesari, Santosh, primary, Schwab, Richard B., primary, Patel, Sandip P., primary, Harismendy, Olivier, primary, Ikeda, Megumi, primary, Parker, Barbara A., primary, and Kurzrock, Razelle, primary
- Published
- 2023
- Full Text
- View/download PDF
46. Data from Genomic Alterations in Circulating Tumor DNA from Diverse Cancer Patients Identified by Next-Generation Sequencing
- Author
-
Schwaederle, Maria, primary, Chattopadhyay, Ranajoy, primary, Kato, Shumei, primary, Fanta, Paul T., primary, Banks, Kimberly C., primary, Choi, In Sil, primary, Piccioni, David E., primary, Ikeda, Sadakatsu, primary, Talasaz, AmirAli, primary, Lanman, Richard B., primary, Bazhenova, Lyudmila, primary, and Kurzrock, Razelle, primary
- Published
- 2023
- Full Text
- View/download PDF
47. Supplementary Table 4 from Genomic Alterations in Circulating Tumor DNA from Diverse Cancer Patients Identified by Next-Generation Sequencing
- Author
-
Schwaederle, Maria, primary, Chattopadhyay, Ranajoy, primary, Kato, Shumei, primary, Fanta, Paul T., primary, Banks, Kimberly C., primary, Choi, In Sil, primary, Piccioni, David E., primary, Ikeda, Sadakatsu, primary, Talasaz, AmirAli, primary, Lanman, Richard B., primary, Bazhenova, Lyudmila, primary, and Kurzrock, Razelle, primary
- Published
- 2023
- Full Text
- View/download PDF
48. Supplementary Table 3 from Genomic Alterations in Circulating Tumor DNA from Diverse Cancer Patients Identified by Next-Generation Sequencing
- Author
-
Schwaederle, Maria, primary, Chattopadhyay, Ranajoy, primary, Kato, Shumei, primary, Fanta, Paul T., primary, Banks, Kimberly C., primary, Choi, In Sil, primary, Piccioni, David E., primary, Ikeda, Sadakatsu, primary, Talasaz, AmirAli, primary, Lanman, Richard B., primary, Bazhenova, Lyudmila, primary, and Kurzrock, Razelle, primary
- Published
- 2023
- Full Text
- View/download PDF
49. Contributors
- Author
-
Abou-Alfa, Ghassan K., primary, Abou Khalil, Jad, additional, Addeo, Pietro, additional, Adsay, N. Volkan, additional, Agarwal, Anil Kumar, additional, Alemi, Farzad, additional, Allen, Peter J., additional, Al-Mukhtar, Ahmed, additional, Aloia, Thomas A., additional, Andersen, Jesper B., additional, Anderson, Christopher D., additional, Arslan-Carlon, Vittoria, additional, Asbun, Horacio J., additional, Aussilhou, Béatrice, additional, Awad, Joseph, additional, Azoulay, Daniel, additional, Bachellier, Philippe, additional, Baker, Talia B., additional, Bamboat, Zubin M., additional, Barkun, Jeffrey Stewart, additional, Bassi, Claudio, additional, Basturk, Olca, additional, Beard, Rachel E., additional, Bedossa, Pierre, additional, Belghiti, Jacques, additional, Bellorin-Marin, Omar, additional, Besselink, Marc G.H., additional, Bilchik, Anton J., additional, Blumgart, Leslie H., additional, Boas, Franz Edward, additional, Brody, Lynn A., additional, Brown, Karen T., additional, Bruix, Jordi, additional, Bruno, David A., additional, Brunt, Elizabeth M., additional, Burns, Justin M., additional, Butturini, Giovanni, additional, Caicedo, Juan Carlos, additional, Callery, Mark P., additional, Calvino, Abdul Saied, additional, Carpenter, Danielle H., additional, Carter, C. Ross, additional, Cauchy, François, additional, Chan, Chung Yip, additional, Chan, See Ching, additional, Chapman, William C., additional, Cherqui, Daniel, additional, Cho, Clifford S., additional, Chung, Jin Wook, additional, Clanton, Jesse, additional, Clary, Bryan Marshall, additional, Cleary, Sean Patrick, additional, Collins, Kelly M., additional, Conneely, John Barry, additional, Connell, Louise C., additional, Corvera, Carlos U., additional, Costa, Guido, additional, Covey, Anne M., additional, Crippin, Jeffrey S., additional, Croome, Kristopher P., additional, Dabbous, Hany, additional, D'Angelica, Michael I., additional, Darcy, Michael D., additional, Davis, Jeremy L., additional, de Jonge, Jeroen, additional, DeMatteo, Ronald P., additional, DePeralta, Danielle K., additional, Desai, Niraj M., additional, de Santibañes, Eduardo, additional, de Santibañes, Martin, additional, Dickson, Euan J., additional, DiMaio, Christopher John, additional, Do, Richard Kinh Gian, additional, Dokmak, Safi, additional, Donati, Marcello, additional, Doyle, M.B. Majella, additional, Dudeja, Vikas, additional, Dunphy, Mark, additional, Earl, Truman M., additional, Ebata, Tomoki, additional, Dika, Imane El, additional, El-Gohary, Yousef, additional, Endo, Itaru, additional, Enestvedt, C. Kristian, additional, Espat, N. Joseph, additional, Ethun, Cecilia G., additional, Fan, Sheung Tat, additional, Fanta, Paul T., additional, Farges, Olivier, additional, Ferrone, Cristina R., additional, Fields, Ryan C., additional, Fischer, Mary, additional, Fisher, Sarah B., additional, Flaherty, Devin C., additional, Fong, Yuman, additional, Friedman, Scott L., additional, Gabr, Ahmed, additional, Galloway, John R., additional, Geller, David A., additional, Gerdes, Hans, additional, Gerst, Scott R., additional, Gittes, George K., additional, Glorioso, Jaime, additional, Gluskin, Jill S., additional, Goh, Brian K.P., additional, Gonzalez, Stevan A., additional, Goodman, Karyn A., additional, Gores, Gregory J., additional, Gotuzzo, Eduardo H., additional, Gouma, Dirk J., additional, Greig, Paul D., additional, Griffin, James F., additional, Halloran, Christopher M., additional, Halpern, Neil A., additional, Hammill, Chet W., additional, Hansen, Paul D., additional, Harding, James J., additional, M. Harrison, Ewen, additional, Hartwig, Werner, additional, Hasegawa, Kiyoshi, additional, Hechtman, Jaclyn F., additional, Heimbach, Julie K., additional, Helton, William S., additional, Hemming, Alan W., additional, Henderson, J. Michael, additional, Hirshberg, Asher, additional, Howe, James R., additional, Hughes, Christopher B., additional, Iacobuzio-Donahue, Christine, additional, Jarnagin, William R., additional, Jenkins, Roger L., additional, Jutric, Zeljka, additional, Kahlert, Christoph, additional, Kallini, Joseph Ralph, additional, Kangrga, Ivan, additional, Karanicolas, Paul J., additional, Katz, Seth S., additional, Katz, Steven C., additional, Kelly, Kaitlyn J., additional, Kemeny, Nancy E., additional, Kennedy, Eugene P., additional, Khalili, Korosh, additional, Khan, Adeel S., additional, Khan, Saboor, additional, Kim, Heung Bae, additional, Kingham, T. Peter, additional, Kirk, Allan D., additional, S. Klimstra, David, additional, Kluger, Michael, additional, Knechtle, Stuart J., additional, Koea, Jonathan B., additional, Kokudo, Norihiro, additional, Koliogiannis, Dionysios, additional, Kooby, David A., additional, Korenblat, Kevin, additional, Krebs, Simone, additional, LaQuaglia, Michael J., additional, LaQuaglia, Michael P., additional, LaRusso, Nicholas F., additional, Laurent, Alexis, additional, Lazaridis, Konstantinos N., additional, Leal, Julie N., additional, Lee, Eliza J., additional, Lee, Major Kenneth, additional, Lee, Ser Yee, additional, Lencioni, Riccardo, additional, Liccioni, Alexandre, additional, Lidsky, Michael E., additional, Lin, Chung-Wei, additional, Linehan, David C., additional, Lopez-Solis, Roberto Carlos, additional, Lowell, Jeffrey A., additional, Madoff, David C., additional, Maggi, Jason, additional, Maithel, Shishir K., additional, Majeed, Ali W., additional, Malfertheiner, Peter, additional, Malleo, Giuseppe, additional, Mao, Shennen A., additional, Marchegiani, Giovanni, additional, Marcos, Luis A., additional, Markmann, James F., additional, Marsh, J. Wallis, additional, Martin, Robert C.G., additional, Matsuyama, Ryusei, additional, Matter, Matthias S., additional, Mattera, Francisco Juan, additional, Maxwell, Jessica E., additional, Mazza, Oscar M., additional, McGilvray, Ian D., additional, McKay, Colin J., additional, McWeeney, Doireann M., additional, Melendez, Jose, additional, Mendelsohn, Robin B., additional, Miller, George, additional, Mönkemüller, Klaus E., additional, Mori, Ryutaro, additional, Moutinho, Vitor, additional, Nagino, Masato, additional, Nagorney, David M., additional, Nagula, Satish, additional, Nakeeb, Attila, additional, Nedredal, Geir I., additional, Neoptolemos, John P., additional, Neuberger, James, additional, Nyberg, Scott L., additional, O'Connor, Rachel, additional, O'Grady, John G., additional, Oldfield, Frances E., additional, Oldhafer, Karl J., additional, Olthoff, Kim M., additional, Orloff, Susan L., additional, Paniccia, Alessandro, additional, Paradis, Valérie, additional, Parks, Rowan W., additional, Pascal, Gérard, additional, Pastores, Stephen M., additional, Pawlik, Timothy M., additional, Pillarisetty, Venu G., additional, Pingpank, James Francis, additional, Pinson, C. Wright, additional, Pitt, Henry Anthony, additional, Pomposelli, James J., additional, Procopio, Fabio, additional, Pucci, Michael J., additional, Qadan, Motaz, additional, Rajkomar, Kheman, additional, Reddy, Srinevas K., additional, Reig, Maria E., additional, Reza, Joseph Arturo, additional, Roberts, John Paul, additional, Robson, Piera Marie Cote, additional, Rocha, Flavio G., additional, Roll, Garrett Richard, additional, Ronnekleiv-Kelly, Sean M., additional, Rosemurgy, Alexander S., additional, Rosen, Charles B., additional, Saldinger, Pierre F., additional, Salem, Riad, additional, Salem, Suhail Bakr, additional, Salvia, Roberto, additional, Sandroussi, Charbel, additional, Sanford, Dominic E., additional, Scatton, Olivier, additional, Schattner, Mark Andrew, additional, Schecter, William Palmer, additional, Schoellhammer, Hans Francis, additional, Schulick, Richard D., additional, Schwartz, Lawrence H., additional, Shah, Kevin N., additional, Shepherd, Ross W., additional, Shimada, Hiroshi, additional, Shimoda, Masafumi, additional, Shindoh, Junichi, additional, Shokouh-Amiri, Hosein, additional, Sicklick, Jason K., additional, Siegelbaum, Robert H., additional, Singh, Gagandeep, additional, Smoot, Rory L., additional, Solomon, Stephen B., additional, Soubrane, Olivier, additional, Spinelli, Nicholas, additional, Stauffer, John A., additional, Stewart, Lygia, additional, Strand, Matthew S., additional, Tabibian, James H., additional, Torzilli, Guido, additional, Trotter, James F., additional, Turcotte, Simon, additional, Turmelle, Yumirle P., additional, Tzimas, Demetrios J., additional, Van Gulik, Thomas, additional, Vannucci, Andrea, additional, Vauthey, Jean-Nicolas, additional, Vetter, Diana, additional, Vilgrain, Valérie, additional, Villamil, Alejandra Maria, additional, Voigt, Louis P., additional, Vollmer, Charles M., additional, Wands, Jack R., additional, Wattacheril, Julia, additional, Weber, Sharon Marie, additional, Weiss, Matthew J., additional, Weitz, Jürgen, additional, Werner, Jens, additional, Winner, Megan, additional, Wong, John, additional, Yang, Dennis, additional, Yarmohammadi, Hooman, additional, Yeo, Charles J., additional, Yeo, Theresa Pluth, additional, Yoon, Chang Jin, additional, Yopp, Adam, additional, Young, D. Owen, additional, Zhao, Kai, additional, Zibari, Gazi B., additional, and Zogopoulos, George, additional
- Published
- 2017
- Full Text
- View/download PDF
50. Molecular pathogenesis of biliary tract cancer
- Author
-
Sicklick, Jason K., primary and Fanta, Paul T., additional
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.