Jon Chung
Foundation Medicine
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Jon Chung.
Annals of Oncology | 2016
Jon Chung; Eric M. Sanford; Adrienne Johnson; Samuel J. Klempner; Alexa B. Schrock; Norma Alonzo Palma; Rachel L. Erlich; Garrett Michael Frampton; Zachary R. Chalmers; Jo-Anne Vergilio; Douglas A. Rubinson; James Sun; Juliann Chmielecki; Roman Yelensky; James Suh; Doron Lipson; Thomas J. George; Julia A. Elvin; P.J. Stephens; V.A. Miller; J.S. Ross; Siraj M. Ali
BACKGROUNDnSquamous cell cancers of the anal canal (ASCC) are increasing in frequency and lack effective therapies for advanced disease. Although an association with human papillomavirus (HPV) has been established, little is known about the molecular characterization of ASCC. A comprehensive genomic analysis of ASCC was undertaken to identify novel genomic alterations (GAs) that will inform therapeutic choices for patients with advanced disease.nnnPATIENTS AND METHODSnHybrid-capture-based next-generation sequencing of exons from 236 cancer-related genes and intronic regions from 19 genes commonly rearranged in cancer was performed on 70 patients with ASCC. HPV status was assessed by aligning tumor sequencing reads to HPV viral genomes. GAs were identified using an established algorithm and correlated with HPV status.nnnRESULTSnSixty-one samples (87%) were HPV-positive. A mean of 3.5 GAs per sample was identified. Recurrent alterations in phosphoinositol-3-kinase pathway (PI3K/AKT/mTOR) genes including amplifications and homozygous deletions were present in 63% of cases. Clinically relevant GAs in genes involved in DNA repair, chromatin remodeling, or receptor tyrosine kinase signaling were observed in 30% of cases. Loss-of-function mutations in TP53 and CDKN2A were significantly enhanced in HPV-negative cases (P < 0.0001).nnnCONCLUSIONSnThis is the first comprehensive genomic analysis of ASCC, and the results suggest new therapeutic approaches. Differing genomic profiles between HPV-associated and HPV-negative ASCC warrants further investigation and may require novel therapeutic and preventive strategies.
Journal of Thoracic Oncology | 2017
Sai-Hong Ignatius Ou; Alexa B. Schrock; Eduard V. Bocharov; Samuel J. Klempner; Carolina Kawamura Haddad; Gary Steinecker; Melissa Lynne Johnson; Barbara J. Gitlitz; Jon Chung; Paulo Vidal Campregher; Jeffrey S. Ross; Philip J. Stephens; Vincent A. Miller; James Suh; Siraj M. Ali; Vamsidhar Velcheti
Introduction: Erb‐b2 receptor tyrosine kinase (HER2) transmembrane domain (TMD) mutations (HER2V659E, HER2G660D) have previously been identified in lung adenocarcinomas, but their frequency and clinical significance is unknown. Methods: We prospectively analyzed 8551 consecutive lung adenocarcinomas using hybrid capture–based comprehensive genomic profiling (CGP) at the request of the individual treating physicians for the purpose of making therapy decisions. Results: We identified 15 cases (0.18%) of HER2 TMD mutations (HER2V659E/D, HER2G660D) through CGP of 8551 lung adenocarcinomas. HER2 TMD mutations were mutually exclusive from HER2 kinase domain mutations and other oncogenic drivers in lung adenocarcinoma. Only two cases with HER2 TMD mutations (13%) had concurrent Erb‐b2 receptor tyrosine kinase 2 gene (HER2) amplification. Structural analysis of HER2 TMD association revealed that mutations at positions V659 and G660 to the highly polar residues glutamic acid, aspartic acid, or arginine should stabilize homodimerization and heterodimerization of HER2 in the active conformation. Treatment with afatinib, a pan‐HER inhibitor, resulted in durable clinical response in three of four patients with lung adenocarcinoma, with two harboring HER2V659E and one with double HER2V659E/G660R mutations. HER2 TMD mutations (V659 and G660) are found in other non‐NSCLC malignancies, and analogous TMD mutations are also found in EGFR, HER3, and HER4. Conclusion: HER2 TMD mutations represent rare but distinct targetable driver mutations in lung adenocarcinoma. CGP capable of detecting diverse HER2 alterations, including HER2 TMD mutations, should be broadly adopted to identify all patients who may benefit from HER2‐targeted therapies.
Annals of Oncology | 2017
Jon Chung; Dean Pavlick; Ryan J. Hartmaier; Alexa B. Schrock; Lauren Young; B Forcier; P Ye; M K Levin; Michael E. Goldberg; Howard A. Burris; A D Hoffman; P.J. Stephens; Garrett Michael Frampton; Doron Lipson; D M Nguyen; Shridar Ganesan; B H Park; Linda T. Vahdat; Brian Leyland-Jones; Tariq I. Mughal; Lajos Pusztai; J O’Shaughnessy; V.A. Miller; J.S. Ross; Siraj M. Ali
BACKGROUNDnGenomic changes that occur in breast cancer during the course of disease have been informed by sequencing of primary and metastatic tumor tissue. For patients with relapsed and metastatic disease, evolution of the breast cancer genome highlights the importance of using a recent sample for genomic profiling to guide clinical decision-making. Obtaining a metastatic tissue biopsy can be challenging, and analysis of circulating tumor DNA (ctDNA) from blood may provide a minimally invasive alternative.nnnPATIENTS AND METHODSnHybrid capture-based genomic profiling was carried out on ctDNA from 254 female patients with estrogen receptor-positive breast cancer. Peripheral blood samples were submitted by clinicians in the course of routine clinical care between May 2016 and March 2017. Sequencing of 62 genes was carried out to a median unique coverage depth of 7503×. Genomic alterations (GAs) in ctDNA were evaluated and compared with matched tissue samples and genomic datasets of tissue from breast cancer.nnnRESULTSnAt least 1GA was reported in 78% of samples. Frequently altered genes were TP53 (38%), ESR1 (31%) and PIK3CA (31%). Temporally matched ctDNA and tissue samples were available for 14 patients; 89% of mutations detected in tissue were also detected in ctDNA. Diverse ESR1 GAs including mutation, rearrangement and amplification, were observed. Multiple concurrent ESR1 GAs were observed in 40% of ESR1-altered cases, suggesting polyclonal origin; ESR1 compound mutations were also observed in two cases. ESR1-altered cases harbored co-occurring GAs in PIK3CA (35%), FGFR1 (16%), ERBB2 (8%), BRCA1/2 (5%), and AKT1 (4%).nnnCONCLUSIONSnGAs relevant to relapsed/metastatic breast cancer management were identified, including diverse ESR1 GAs. Genomic profiling of ctDNA demonstrated sensitive detection of mutations found in tissue. Detection of amplifications was associated with ctDNA fraction. Genomic profiling of ctDNA may provide a complementary and possibly alternative approach to tissue-based genomic testing for patients with estrogen receptor-positive metastatic breast cancer.Abstract Background Genomic changes that occur in breast cancer during the course of disease have been informed by sequencing of primary and metastatic tumor tissue. For patients with relapsed and metastatic disease, evolution of the breast cancer genome highlights the importance of using a recent sample for genomic profiling to guide clinical decision-making. Obtaining a metastatic tissue biopsy can be challenging, and analysis of circulating tumor DNA (ctDNA) from blood may provide a minimally invasive alternative. Patients and methods Hybrid capture-based genomic profiling was carried out on ctDNA from 254 female patients with estrogen receptor-positive breast cancer. Peripheral blood samples were submitted by clinicians in the course of routine clinical care between May 2016 and March 2017. Sequencing of 62 genes was carried out to a median unique coverage depth of 7503×. Genomic alterations (GAs) in ctDNA were evaluated and compared with matched tissue samples and genomic datasets of tissue from breast cancer. Results At least 1 GA was reported in 78% of samples. Frequently altered genes were TP53 (38%), ESR1 (31%) and PIK3CA (31%). Temporally matched ctDNA and tissue samples were available for 14 patients; 89% of mutations detected in tissue were also detected in ctDNA. Diverse ESR1 GAs including mutation, rearrangement and amplification, were observed. Multiple concurrent ESR1 GAs were observed in 40% of ESR1-altered cases, suggesting polyclonal origin; ESR1 compound mutations were also observed in two cases. ESR1-altered cases harbored co-occurring GAs in PIK3CA (35%), FGFR1 (16%), ERBB2 (8%), BRCA1/2 (5%), and AKT1 (4%). Conclusions GAs relevant to relapsed/metastatic breast cancer management were identified, including diverse ESR1 GAs. Genomic profiling of ctDNA demonstrated sensitive detection of mutations found in tissue. Detection of amplifications was associated with ctDNA fraction. Genomic profiling of ctDNA may provide a complementary and possibly alternative approach to tissue-based genomic testing for patients with estrogen receptor-positive metastatic breast cancer.
European Urology | 2018
Sumanta K. Pal; Siraj M. Ali; Evgeny Yakirevich; Daniel M. Geynisman; Jose A. Karam; Julia A. Elvin; Garrett Michael Frampton; Xuan Huang; Douglas I. Lin; Mark Rosenzweig; Doron Lipson; Philip J. Stephens; Jeffrey S. Ross; Vincent A. Miller; Neeraj Agarwal; Brian Shuch; Toni K. Choueiri; Jon Chung
BACKGROUNDnPapillary renal cell carcinoma (PRCC) is a rare subset of RCC. The Cancer Genome Atlas (TCGA) data largely reflect localized disease, and there are limited data for advanced PRCC.nnnOBJECTIVEnTo characterize the frequency of genomic alterations (GAs) in patients with advanced PRCC for whom comprehensive genomic profiling (CGP) was performed in the context of routine clinical care.nnnDESIGN, SETTING, AND PARTICIPANTSnFormalin-fixed, paraffin-embedded tissue was obtained for 169 consecutive patients with confirmed PRCC. DNA was extracted and comprehensive genomic profiling was performed in a certified central laboratory.nnnMEASUREMENTSnHybrid-capture, adaptor ligation-based libraries of up to 315 genes were sequenced to a median coverage of 648×. All classes of GAs were identified, including substitutions, insertions/deletions, copy number alterations, and rearrangements.nnnRESULTS AND LIMITATIONSnFrom 169 patients, either primary tumor tissue (102 patients, 60%) or metastatic tissue (67 patients, 40%) was collected. In patients with type 1 PRCC, commonly altered genes were MET (33%; 8 activating mutations, 5 amplifications at >6 copies), TERT (30%), CDKN2A/B (13%), and EGFR (8%). In patients with type 2 PRCC, commonly altered genes were CDKN2A/B (18%), TERT (18%), NF2 (13%), and FH (13%); MET GAs (5 mutations, 3 amplifications) were observed in 7% of type 2 cases. Notable differences from TCGA data include higher frequencies of MET, NF2, and CDKN2A/B GAs, association of alterations in SWI/SNF complex genes with type 2 PRCC, and observation of frequent CDKN2A/B alterations in both type 1 and type 2 disease.nnnCONCLUSIONSnBoth the current study and the TCGA experience represent similarly sized cohorts of patients with PRCC. Key differences in GA frequency probably underscore the marked difference in stage distribution between these data sets. These results may inform planned precision medicine trials for metastatic PRCC.nnnPATIENT SUMMARYnPapillary renal cell carcinoma (PRCC) is a rare subtype of kidney cancer, and understanding of the biology of advanced PRCC is limited. This report highlights some of the unique biologic features of PRCC that may inform on future use of targeted therapies for the treatment of metastatic disease.
Breast Cancer Research and Treatment | 2017
Sahar Nozad; Christine E. Sheehan; Julia A. Elvin; Jo-Anne Vergilio; James Suh; Shakti Ramkissoon; Alexa B. Schrock; Kim M. Hirshfield; Nadia Dawn Ali; Shridar Ganesan; Siraj M. Ali; Vincent A. Miller; Philip J. Stephens; Jeffrey S. Ross; Jon Chung
PurposeMalignant phyllodes tumors (MPT) are exceptionally rare, and the genomic drivers of these tumors are still being elucidated. We performed comprehensive genomic profiling (CGP) of MPT to identify genomic alterations that will inform approaches to targeted therapy for patients with MPT, including relapsed, refractory, and metastatic disease.MethodsDNA was extracted from formalin-fixed, paraffin-embedded samples from 24 consecutive patient cases of MPT. CGP was performed using a hybrid capture, adaptor ligation-based next generation sequencing assay to a high, uniform coverage (mean, 582×). Tumor mutational burden (TMB) was calculated from a minimum of 1.14xa0Mb of sequenced DNA as previously described and reported as mutations/Mb. The results were analyzed for all classes of genomic alterations, including short variants (SV; base substitutions, small insertions, and deletions), rearrangements, and copy number changes, including amplifications and homozygous deletions.ResultsThe 24 cases of MPT included 15 patients with localized and 9 with metastatic disease. The median TMB was 2.7 mut/Mb, and no cases had a TMBxa0>xa010 mut/Mb. 20 out of 24 cases were evaluable for microsatellite status, and all were microsatellite stable. The most commonly mutated genes were TP53 (58.3%), TERT-promoter (57.9%), NF1 (45.8%), MED12 (45.8%), CDKN2A/B (33.3%), and MLL2 (33.3%). Targetable kinase fusions including KIAA1549-BRAF or FGFR3-TACC3 were identified in 2/24 (8.3%) tumors.ConclusionsThis study identifies clinically relevant genomic alterations that suggest novel targeted therapy approaches for patients with MPT.
Annals of Oncology | 2018
Ryan J. Hartmaier; Sally E. Trabucco; Nolan Priedigkeit; Jon Chung; C. A. Parachoniak; P. Vanden Borre; S. Morley; Mark Rosenzweig; Michael E. Goldberg; James Suh; Siraj M. Ali; Jeffrey S. Ross; Brian Leyland-Jones; B. Young; C. Williams; Ben Ho Park; M. Tsai; Barbara Haley; Julio Peguero; R. D. Callahan; I. Sachelarie; J. Cho; J. M. Atkinson; Amir Bahreini; Alison M. Nagle; S. L. Puhalla; Rebecca J. Watters; Z. Erdogan-Yildirim; L. Cao; Steffi Oesterreich
Abstract Background Estrogen receptor-positive (ER-positive) metastatic breast cancer is often intractable due to endocrine therapy resistance. Although ESR1 promoter switching events have been associated with endocrine-therapy resistance, recurrent ESR1 fusion proteins have yet to be identified in advanced breast cancer. Patients and methods To identify genomic structural rearrangements (REs) including gene fusions in acquired resistance, we undertook a multimodal sequencing effort in three breast cancer patient cohorts: (i) mate-pair and/or RNAseq in 6 patient-matched primary-metastatic tumors and 51 metastases, (ii) high coverage (>500×) comprehensive genomic profiling of 287–395 cancer-related genes across 9542 solid tumors (5216 from metastatic disease), and (iii) ultra-high coverage (>5000×) genomic profiling of 62 cancer-related genes in 254 ctDNA samples. In addition to traditional gene fusion detection methods (i.e. discordant reads, split reads), ESR1 REs were detected from targeted sequencing data by applying a novel algorithm (copyshift) that identifies major copy number shifts at rearrangement hotspots. Results We identify 88 ESR1 REs across 83 unique patients with direct confirmation of 9 ESR1 fusion proteins (including 2 via immunoblot). ESR1 REs are highly enriched in ER-positive, metastatic disease and co-occur with known ESR1 missense alterations, suggestive of polyclonal resistance. Importantly, all fusions result from a breakpoint in or near ESR1 intron 6 and therefore lack an intact ligand binding domain (LBD). In vitro characterization of three fusions reveals ligand-independence and hyperactivity dependent upon the 3′ partner gene. Our lower-bound estimate of ESR1 fusions is at least 1% of metastatic solid breast cancers, the prevalence in ctDNA is at least 10× enriched. We postulate this enrichment may represent secondary resistance to more aggressive endocrine therapies applied to patients with ESR1 LBD missense alterations. Conclusions Collectively, these data indicate that N-terminal ESR1 fusions involving exons 6–7 are a recurrent driver of endocrine therapy resistance and are impervious to ER-targeted therapies.
Clinical Cancer Research | 2018
Alexa B. Schrock; Dean Pavlick; Samuel J. Klempner; Jon Chung; Brady Forcier; Allison Welsh; Lauren Young; Brian Leyland-Jones; Rodolfo Bordonii; Richard D. Carvajal; Joseph Chao; Razelle Kurzrock; Jason K. Sicklick; Jeffrey S. Ross; Philip J. Stephens; Craig Devoe; Fadi S. Braiteh; Siraj M. Ali; Vincent A. Miller
Purpose: Genomic profiling of tumor biopsies from advanced gastrointestinal and anal cancers is increasingly used to inform treatment. In some cases, tissue biopsy can be prohibitive, and we sought to investigate whether analysis of blood-derived circulating tumor DNA (ctDNA) may provide a minimally invasive alternative. Experimental Design: Hybrid capture–based genomic profiling of 62 genes was performed on blood-based ctDNA from 417 patients with gastrointestinal carcinomas to assess the presence of genomic alterations (GA) and compare with matched tissue samples. Results: Evidence of ctDNA was detected in 344 of 417 samples (82%), and of these, ≥1 reportable GA was detected in 89% (306/344) of samples. Frequently altered genes were TP53 (72%), KRAS (35%), PIK3CA (14%), BRAF (8%), and EGFR (7%). In temporally matched ctDNA and tissue samples available from 25 patients, 86% of alterations detected in tissue were also detected in ctDNA, including 95% of short variants, but only 50% of amplifications. Conversely, 63% of alterations detected in ctDNA were also detected in matched tissue. Examples demonstrating clinical utility are presented. Conclusions: Genomic profiling of ctDNA detected potentially clinically relevant GAs in a significant subset of patients with gastrointestinal carcinomas. In these tumor types, most alterations detected in matched tissue were also detected in ctDNA, and with the exception of amplifications, ctDNA sequencing routinely detected additional alterations not found in matched tissue, consistent with tumor heterogeneity. These results suggest feasibility and utility of ctDNA testing in advanced gastrointestinal cancers as a complementary approach to tissue testing, and further investigation is warranted. Clin Cancer Res; 24(8); 1881–90. ©2018 AACR.
European urology focus | 2018
Andrea Necchi; Gennady Bratslavsky; Robert John Corona; Jon Chung; Sherri Z. Millis; Julia A. Elvin; Jo-Anne Vergilio; James Suh; Shakti Ramkissoon; Eric Allan Severson; Sugganth Daniel; Jonathan Keith Killian; Siraj M. Ali; Alexa B. Schrock; Prasanth Reddy; Vincent A. Miller; Allison Welsh; Jeffrey S. Ross
BACKGROUNDnAlthough both seminomatous and nonseminomatous testicular germ cell tumors (TGCTs) have favorable outcomes with chemotherapy, a subset is chemorefractory, and novel therapeutic options are needed.nnnOBJECTIVEnTo molecularly characterize chemotherapy-refractory TGCTs.nnnDESIGN, SETTING, AND PARTICIPANTSnArchival tissues from 107 chemotherapy-treated and relapsed TGCT patients (23 seminomas; 84 nonseminomas) underwent hybrid-capture-based genomic profiling to evaluate four classes of genomic alterations (GAs). Tumor mutational burden (TMB) and microsatellite instability (MSI) were also measured.nnnINTERVENTIONnGenomic profiling on tumor samples from chemotherapy-refractory TGCTs.nnnOUTCOME MEASUREMENTS AND STATISTICAL ANALYSISnDescriptive analyses and differences between seminoma and nonseminoma subgroups were reported.nnnRESULTS AND LIMITATIONSnThe mean GA/tumor was 2.9 for seminomas and 4.0 for nonseminomas (p=0.04). KRAS alterations (mainly amplifications) were the most common GAs at the single-gene level (47.8% of seminomas and 51.2% of nonseminomas). RAS-RAF pathway (56.5% vs 52.3%) and cell-cycle pathway (52.2% vs 56.0%) were the most common GA classes in seminomas and nonseminomas, respectively. Receptor tyrosine kinase pathway and PI3K pathway GAs were more frequent in seminomas (p=0.02). Median TMB was 1.8 mutations/Mb for seminomas and 2.7 mutations/Mb for nonseminomas (p=0.098), and MSI-high status was found in one nonseminoma only (1.2%). A lack of clinical outcome correlation is a limitation of the present analyses.nnnCONCLUSIONSnIn chemotherapy-refractory TGCTs, trials with agents targeting the KRAS pathway may be pursued due to the high frequency of KRAS GAs. Overall, the GAs found in refractory seminomas and nonseminomas differ significantly. Considering the frequency of high TMB or MSI-high status, immunotherapy may benefit a small subset of nonseminomas.nnnPATIENT SUMMARYnTesticular cancers that are resistant to or relapse after standard chemotherapy may harbor genomic alterations that are potentially druggable, particularly in the clinical trial setting, and genomic profiling can guide clinical research and disclose therapeutic opportunities for these patients.
The Journal of Molecular Diagnostics | 2018
Travis A. Clark; Jon Chung; Mark Kennedy; Jason D. Hughes; Niru Chennagiri; Daniel S. Lieber; Bernard Fendler; Lauren Young; Mandy Zhao; Michael Coyne; Virginia Breese; Geneva Young; Amy Donahue; Dean Pavlick; Alyssa Tsiros; Tim Brennan; Shan Zhong; Tariq I Mughal; Mark Bailey; Jie He; Steven Roels; Garrett Michael Frampton; Jill M. Spoerke; Steven Gendreau; Mark R. Lackner; Erica Schleifman; Eric Peters; Jeffrey S. Ross; Siraj M. Ali; Vincent A. Miller
Genomic profiling of circulating tumor DNA derived from cell-free DNA (cfDNA) in blood can provide a noninvasive method for detecting genomic biomarkers to guide clinical decision making for cancer patients. We developed a hybrid capture–based next-generation sequencing assay for genomic profiling of circulating tumor DNA from blood (FoundationACT). High-sequencing coverage and molecular barcode–based error detection enabled accurate detection of genomic alterations, including short variants (base substitutions, short insertions/deletions) and genomic re-arrangements at low allele frequencies (AFs), and copy number amplifications. Analytical validation was performed on 2666 reference alterations. The assay achieved >99% overall sensitivity (95% CI, 99.1%–99.4%) for short variants at AF >0.5%, >95% sensitivity (95% CI, 94.2%–95.7%) for AF 0.25% to 0.5%, and 70% sensitivity (95% CI, 68.2%–71.5%) for AF 0.125% to 0.25%. No false positives were detected in 62 samples from healthy volunteers. Genomic alterations detected by FoundationACT demonstrated high concordance with orthogonal assays run on the same clinical cfDNA samples. In 860 routine clinical FoundationACT cases, genomic alterations were detected in cfDNA at comparable frequencies to tissue; for the subset of cases with temporally matched tissue and blood samples, 75% of genomic alterations and 83% of short variant mutations detected in tissue were also detected in cfDNA. On the basis of analytical validation results, FoundationACT has been approved for use in our Clinical Laboratory Improvement Amendments–certified/College of American Pathologists–accredited/New York State–approved laboratory.
European Urology | 2018
Andrea Necchi; Patrizia Giannatempo; Daniele Raggi; Luigi Mariani; Maurizio Colecchia; Elena Farè; Francesco Monopoli; Giuseppina Calareso; Siraj M. Ali; Jeffrey S. Ross; Jon Chung; Roberto Salvioni
There is growing interest in the potential antitumor effects of immune checkpoint inhibitors targeting the PD-1/PD-L1 pathway. Data from two independent groups have demonstrated that the majority of patients with germ cell tumors (GCTs) express PD-L1 [1,2]. Furthermore, our analysis of the Foundation Medicine database revealed that a small subset of nonseminomatous GCTs featured a high tumor mutation burden (TMB) or microsatellite instability (MSI) that could make these tumors sensitive to immune checkpoint inhibitor therapy [3]. APACHE (NCT03081923) is an open-label randomized phase 2 study. Patients were randomized 1:1 to receive 1500 mg of durvalumab intravenously (arm A) or 1500 mg of durvalumab plus 75 mg of tremelimumab intravenously (arm B) for four cycles followed by durvalumab alone. Treatment was administered every 4 wk in both study arms until disease progression (PD), unacceptable toxicity onset, or a maximum of 12 mo was achieved. The primary endpoint was the objective response rate (according to Response Evaluation Criteria in Solid Tumor [RECIST] version 1.1). The total sample size was divided into a three-stage design. In stage 1, each arm is terminated if no responses are observed in 11 patients (details on the study design are provided in the Supplementary material). Biomarker analyses of the available pretherapy tumor samples included PD-L1 expression (Ventana SP142 assay, details provided in the Supplementary material) and genomic sequencing (FoundationONE assay, Foundation Medicine, Cambridge, MA, USA). From February 2017 to April 2018, 22 patients were randomized at a single center (11 per arm, Supplementary Table 1). The median follow-up (calculated using the